WO2008111990A1 - Analyse de cellules rares par division d'échantillon et utilisation de marqueurs d'adn - Google Patents

Analyse de cellules rares par division d'échantillon et utilisation de marqueurs d'adn Download PDF

Info

Publication number
WO2008111990A1
WO2008111990A1 PCT/US2007/071148 US2007071148W WO2008111990A1 WO 2008111990 A1 WO2008111990 A1 WO 2008111990A1 US 2007071148 W US2007071148 W US 2007071148W WO 2008111990 A1 WO2008111990 A1 WO 2008111990A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
rare
subsamples
patient
Prior art date
Application number
PCT/US2007/071148
Other languages
English (en)
Inventor
Daniel Schoemaker
Martin Fuchs
Neil X. Krueger
Mehmet Toner
Darren Gray
Ravi Kapur
Zihua Wang
Original Assignee
Cellpoint Diagnostics, Inc.
Living Microsystems, Inc.
The General Hospital Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cellpoint Diagnostics, Inc., Living Microsystems, Inc., The General Hospital Corporation filed Critical Cellpoint Diagnostics, Inc.
Publication of WO2008111990A1 publication Critical patent/WO2008111990A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/40Concentrating samples
    • G01N1/405Concentrating samples by adsorption or absorption
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume, or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N2015/1006Investigating individual particles for cytology
    • G01N2015/1029
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/38Pediatrics
    • G01N2800/385Congenital anomalies

Definitions

  • Analysis of specific cells can give insight mto a variety of diseases. These analyses can provide non- invasive tests for detection, diagnosis and prognosis of diseases such as cancer or fetal disorders, thereby eliminating the risk of invasive diagnosis.
  • diseases such as cancer or fetal disorders
  • current prenatal diagnosis such as amniocentesis and chorionic villus sampling (CVS)
  • CVS chorionic villus sampling
  • the rate of miscarriage for pregnant women undergoing amniocentesis is increased by 0.5-1%, and that figure is slightly higher for CVS.
  • ultrasonography is used to determine congenital defects involving neural tube defects and limb abnormalities, but such methods are limited to time periods after fifteen weeks of gestation and present unreliable results.
  • the presence of fetal cells within the blood of pregnant women offers the opportunity to develop a prenatal diagnostic that replaces amniocentesis and thereby eliminates the risk of today's invasive diagnostics.
  • fetal cells represent a small number of cells against the background of a large number of maternal cells in the blood which make the analysis time consuming and prone to error.
  • early detection is of paramount importance. Cancer is a disease marked by the uncontrolled proliferation of abnormal cells. In normal tissue, cells divide and organize within the tissue in response to signals from surrounding cells.
  • Cancer cells do not respond in the same way to these signals, causing them to proliferate and, in many organs, form a tumor. As the growth of a tumor continues, genetic alterations may accumulate, manifesting as a more aggressive growth phenorype of the cancer cells If left untreated, metastasis, the spread of cancer cells to distant areas of the body by way of the lymph system or bloodstream, may ensue. Metastasis results in the formation of secondary tumors at multiple sites, damaging healthy tissue. Most cancer death is caused by such secondary tumors.
  • the methods of the present invention allow for the detection of fetal cells and fetal abnormalities when fetal cells are mixed with a population of maternal cells, even when the maternal cells dominate the mixture.
  • the methods of the present invention can also be utilized to detect, diagnose, or prognose cancer.
  • the present invention relates to methods for the detection of fetal cells or cancer cells in a mixed sample.
  • the present invention provides methods for determining fetal abnormalities in a sample comprising fetal cells that are mixed with a population of maternal cells.
  • determining the presence of fetal cells and fetal abnormalities comprises labeling one or more regions of genomic DNA in each cell from a mixed sample comprising at least one fetal cell with different labels wherein each label is specific to each cell.
  • the genomic DNA to be labeled comprises one or more polymorphisms, particularly STRs or SNPs
  • the methods of the invention allow for simultaneously detecting the presence of fetal cells and fetal abnormalities when fetal cells are mixed with a population of maternal cells, even when the maternal cells dominate the mixture.
  • the sample is enriched to contain at least one fetal and one non fetal cell, and in other embodiments, the cells of the enriched population can be divided between two or more discrete locations that can be used as addressable locations. Examples of addressable locations include wells, bins, sieves, pores, geometric sites, matrixes, membranes, electric traps, gaps or obstacles.
  • the methods comprise labeling one or more regions of genomic DNA in each cell in the enriched sample with different labels, wherein each label is specific to each cell, and quantifying the labeled DNA regions.
  • the labeling methods can comprise adding a unique tag sequence for each cell in the mixed sample.
  • the unique tag sequence identifies the presence or absence of a DNA polymorphism in each cell from the mixed sample.
  • Labels are added to the cells/DNA using an amplification reaction, which can be performed by PCR methods. For example, amplification can be achieved by multiplex PCR. In some embodiments, a further PCR amplification is performed using nested primers for the genomic DNA region(s).
  • the DNA regions can be amplified prior to being quantified.
  • the labeled DNA can be quantified using sequencing methods, which, in some embodiments, can precede amplifying the DNA regions.
  • the amplified DNA region(s) can be analyzed by sequencing methods. For example, ultra deep sequencing can be used to provide an accurate and quantitative measurement of the allele abundances for each STR or SNP.
  • quantitative genotyping can be used to declare the presence of fetal cells and to determine the copy numbers of the fetal chromosomes. Preferably, quantitative genotyping is performed using molecular inversion probes.
  • the invention also relates to methods of identifying cells from a mixed sample with non-maternal genomic DNA and identifying said cells with non-maternal genomic DNA as fetal cells. In some embodiments, the ratio of maternal to paternal alleles is compared on the identified fetal cells in the mixed sample. [0012] In one embodiment, the invention provides for a method for determining a fetal abnormality in a maternal sample that comprises at least one fetal and one non fetal cell. The sample can be enriched to contain at least one fetal cell, and the enriched maternal sample can be arrayed into a plurality of discrete sites. In some embodiments, each discrete site comprises no more than one cell.
  • the invention comprises labeling one or more regions of genomic DNA from the arrayed samples using primers that are specific to each DNA region or location, amplifying the DNA region(s), and quantifying the labeled DNA region.
  • the labeling of the DNA region(s) can comprise labeling each region with a unique tag sequence, which can be used to identify the presence or absence of a DNA polymorphism on arrayed cells and the distinct location of the cells.
  • the step of determining can comprise identifying non-maternal alleles at the distinct locations, which can result from comparing the ratio of maternal to paternal alleles at the location.
  • the method of identifying a fetal abnormality in an arrayed sample can further comprise amplifying the genomic DNA regions.
  • the genomic DNA regions can comprise one or more polymorphisms, e.g., STRs and SNPs, which can be amplified using PCR methods including multiplex PCR. An additional amplification step can be performed using nested primers.
  • the amplified DNA region(s) can be analyzed by sequencing methods.
  • ultra deep sequencing can be used to provide an accurate and quantitative measurement of the allele abundances for each STR or SNP.
  • quantitative genotyping can be used to declare the presence of fetal cells and to determine the copy numbers of the fetal chromosomes.
  • quantitative genotyping is performed using molecular inversion probes.
  • the invention provides methods for diagnosing a cancer and giving a prognosis by obtaining and enriching a blood sample from a patient for epithelial cells, splitting the enriched sample into discrete locations, and performing one or more molecular and/or morphological analyses on the enriched and split sample.
  • the molecular analyses can include detecting the level of expression or a mutation of gene disclosed in Figure 10.
  • the method comprises performing molecular analyses on EGFR, EpCAM, GA733-2, MUC-I, HER-2, or Claudin-7 in each arrayed cell.
  • the morphological analyses can include identifying, quantifying and /or characterizing mitochondrial DNA, telomerase, or nuclear matrix proteins.
  • morphological analyses include staining rare cells and imaging the stained rare cells using bright field microscopy, e.g., to determine cell size, cell shape, nuclear size, nuclear shape, the ratio of cytoplasmic to nuclear volume, etc.
  • the sample can be enriched for epithelial cells by at least 10,000 fold, and the diagnosis and prognosis can be provided prior to treating the patient for the cancer.
  • the blood samples are obtained from a patient at regular intervals such as daily, or every 2, 3 or 4 days, weekly, bimonthly, monthly, bi-yearly or yearly.
  • the step of enriching a patient's blood sample for epithelial cells involves flowing the sample through a first array of obstacles that selectively directs cells that are larger than a predetermined size to a first outlet and cells that are smaller than a predetermined size to a second outlet.
  • the sample can be subjected to further enrichment by flowing the sample through a second array of obstacles, which can be coated with antibodies that selectively bind to white blood cells or epithelial cells.
  • the obstacles of the second array can be coated with anti-EpCAM antibodies.
  • Splitting the sample of cells of the enriched population can comprises splitting the enriched sample to locate individual cells at discrete sites that can be addressable sites. Examples of addressable locations include wells, bins, sieves, pores, geometric sites, matrixes, membranes, electric traps, gaps or obstacles.
  • kits comprising devices for enriching the sample and the devices and reagents needed to perform the genetic analysis.
  • kits may contain the arrays for size-based separation, reagents for uniquely labeling the cells, devices for splitting the cells into individual addressable locations and reagents for the genetic analysis.
  • the present invention provides a method for diagnosing or prognosing cancer in a patient.
  • the method comprises splitting a rare cell-enriched biological sample, obtained at a time point from the patient, into a plurality of subsamples and performing a molecular analysis or a morphological analysis on one or more subsamples in the plurality of subsamples, where performing a molecular analysis or a morphological analysis on one or more subsamples in said plurality of subsamples, where ten percent or more of the total number of cells in at least one of the one or more subsamples are rare cells.
  • a cancer diagnosis or prognosis for the patient is then determined based on the molecular analysis or the morphological analysis.
  • the method includes determining the fraction of subsamples that comprise one or more rare cells.
  • the plurality of subsamples is at least 10 subsamples.
  • One or more of the rare cells contained in one or more subsamples in the plurality of subsamples can be an epithelial cell, a circulating tumor cell, an endothelial cell, or a stem cell.
  • one or more of the rare cells can be an epithelial cell.
  • the rare cell-enriched biological sample can be a rare cell-enriched blood sample.
  • At least one of the plurality of subsamples can comprise about one to ten rare cells.
  • At least one of the plurality of subsamples can comprise about one to five rare cells.
  • Each of the plurality of subsamples can contain about one to five rare cells.
  • Each of the plurality of subsamples can contain one rare cell.
  • the method further comprises determining a total number of rare cells in the rare cell enriched biological sample.
  • the method further comprises splitting into a plurality of subsamples one or more rare cell enriched biological samples obtained from the patient at one or more time points subsequent to the time point.
  • the time points can occur at an interval between one day and one year subsequent to the time point.
  • the time points can occur at a regular time interval subsequent to the time point, for example, two weeks, one month, two months, three months, six months, or one year.
  • the rare cell-enriched biological sample can be obtained from a patient who had not undergone cancer therapy or from a patient who had undergone cancer therapy.
  • the rare cell-enriched biological sample can be obtained by rare cell immunoaffmity separation of a biological sample from the patient.
  • the rare cell immunoaffinity separation can include flowing the biological sample from the patient through an array of obstacles coated with one or more antibodies that selectively bind to the rare cells.
  • the one or more antibodies can comprise anti-
  • the biological sample from the patient can be flowed through an array of obstacles that selectively directs cells larger than a predetermined size to a first outlet and cells smaller than a predetermined size to a second outlet.
  • the rare cell-enriched biological sample can be obtained by size based separation of rare cells present in a biological sample from the patient.
  • the size-based separation of rare cells can include flowing a biological sample from the patient through an array of obstacles that deflect particles based on hydrodynamic size.
  • the biological sample from the patient can be flowed through an array of obstacles coated with antibodies that selectively bind to rare cells.
  • the rare cell-enriched biological sample can be enriched in rare cells by at least 100 fold.
  • At least one of the subsamples in the plurality of subsamples can occupy a discrete site.
  • the discrete site can be a well.
  • the discrete site can be addressable.
  • the splitting of a rare cell-enriched biological sample can generate multiple subsamples substantially at the same time.
  • the splitting can generate at least 14 of the subsamples at the same time.
  • the splitting can be automated. [0027]
  • the molecular analysis can comprise detecting the presence or absence of a mutation in a gene identified in
  • the gene can be an EGFR gene.
  • the mutation can occur in any of exons 18-21 of the EGFR gene.
  • the molecular analysis can comprise detecting expression of a gene identified in Figure 10.
  • the gene can be EGFR, EGF, EpCAM, GA733-2, MUC-I, HER-2, or Claudin-7.
  • the gene can be EpCAM.
  • the gene can be EGFR or EGF.
  • the level of expression of the gene can be determined.
  • the molecular analysis can comprise analyzing mitochondrial DNA, telomerase, a nuclear matrix protein, or a microRNA.
  • the morphological analysis can comprise staining and performing bright-field imaging of the one or more rare cells.
  • the molecular analysis can comprise amplifying one or more genomic sequences from the one or more rare cells to generate genomic amplicons.
  • the amplifying can comprise tagging the one or more genomic sequences to generate tagged genomic amplicons.
  • the tagged genomic amplicons can be locator elements.
  • the amplifying can be followed by ultra deep sequence analysis.
  • the amplifying can also be followed by quantitative genotyping.
  • the quantitative genotyping can further comprise determining a genomic sequence copy number.
  • the quantitative genotyping can be performed using one or more molecular inversion probes.
  • the amplifying can comprise performing quantitative PCR, quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RT-PCR), single cell PCR, restriction fragment length polymorphism PCR
  • PCR-RFLP PCR-RFLP/RT-PCR-RFLP
  • hot start PCR in situ polony PCR
  • in situ rolling circle amplification RCA
  • bridge PCR picotiter PCR
  • emulsion PCR ligase chain reaction (LCR)
  • transcription amplification self-sustained sequence replication, selective amplification of target polynucleotide sequences, consensus sequence primed polymerase chain reaction (CP-PCR), arbitrarily primed polymerase chain reaction (AP-PCR), degenerate oligonucleotide-primed PCR (DOP-PCR), or nucleic acid sequence based amplification (NASBA).
  • the molecular analysis can comprise performing a molecular beacon assay on the one or more rare cells.
  • the present invention further provides a method for diagnosing or prognosing cancer in a patient.
  • the method comprises: (i) enriching a biological sample, obtained at a time point from the patient, for rare cells to obtain a rare cell-enriched biological sample; (ii) splitting the rare cell-enriched biological sample obtained from the patient at a time point into a plurality of subsamples; and (iii) performing a molecular analysis or a morphological analysis on one or more rare cells contained in one or more subsamples in the plurality of subsamples.
  • the cancer diagnosis or prognosis for the patient is determined based on the molecular analysis or the morphological analysis.
  • the plurality of subsamples can comprise at least 10 subsamples.
  • One or more rare cells contained in one or more subsamples in the plurality of subsamples can comprise an epithelial cell, a circulating tumor cell, an endothelial cell, or a stem cell.
  • one or more rare cells contained in one or more subsamples in the plurality of subsamples can be an epithelial cell.
  • the biological sample can be a blood sample.
  • the biological sample can be treated with a stabilizer, a preservative, a fixant, an anti-apoptotic reagent, an anti-coagulation reagent, an anti-thrombotic reagent, a buffering reagent, an osmolality regulating reagent, a pH regulating reagent, or a cross-linking reagent.
  • the biological sample can be treated with a cell viability stain or a cell inviability stain. At least one of the subsamples in the plurality of subsamples can comprise about one to ten rare cells.
  • At least one of the subsamples in the plurality of subsamples can comprise about one to five rare cells.
  • Each of the subsamples in the plurality of subsamples can comprise about one to five rare cells.
  • the subsample can comprise one rare cell.
  • the method for diagnosing or prognosing cancer in a patient can further comprise repeating steps (i) to (iii), such that one or more biological samples are obtained at one or more time points subsequent to the time point.
  • the one or more time points can occur at an interval between one day and one year subsequent to the time point.
  • the one or more time points can occur at a regular time interval subsequent to the time point, for example, two weeks, one month, two months, three months, six months, or one year.
  • the biological sample is obtained from a patient who had not undergone cancer therapy or from a patient who had undergone cancer therapy.
  • the enriching can comprise performing rare cell immunoaffinity separation on the biological sample.
  • the rare cell immunoaffinity separation can comprise flowing the biological sample through an array of obstacles coated with one or more antibodies that selectively bind to the rare cells.
  • One or more antibodies can comprise anti-EpCAM antibodies.
  • the rare cell-enriched biological sample can be enriched in rare cells by at least 100 fold. At least one of the subsamples in the plurality of subsamples can occupy a discrete site. The discrete site can be addressable.
  • splitting of the rare cell-enriched biological sample obtained from the patient can generate multiple subsamples substantially at the same time, for example, at least 14 of the subsamples substantially at the same time.
  • the splitting can be automated.
  • molecular analysis can comprise detecting the presence or absence of a mutation in a gene identified in Figure 10.
  • the gene can be an EGFR gene.
  • the mutation can occur in any of exons 18-21 of the EGFR gene.
  • the molecular analysis can comprise detecting expression of a gene identified in Figure 10.
  • the gene can be EGFR, EGF, EpCAM, GA733-2, MUC-I, HER-2, or Claudin 7. In one embodiment, the gene can be EpCAM. In other embodiments, the gene can be EGFR or EGF.
  • the level of expression of the gene can be determined.
  • the molecular analysis can comprise analyzing mitochondrial DNA, telomerase, a nuclear matrix protein, or a microRNA.
  • the molecular analysis can comprise performing a molecular beacon assay on the one or more rare cells.
  • the morphological analysis can comprise staining and performing bright- field imaging of the one or more rare cells.
  • the molecular analysis can comprise amplifying one or more genomic sequences from the one or more rare cells to generate genomic amplicons.
  • the amplifying can comprise tagging the one or more genomic sequences to generate tagged genomic amplicons.
  • the tagged genomic amplicons can comprise locator elements.
  • the amplifying can be followed by ultra deep sequence analysis.
  • the amplifying can be followed by quantitative genotyping.
  • the quantitative genotyping can comprise determining a genomic sequence copy number.
  • the quantitative genotyping can be performed using one or more molecular inversion probes.
  • the amplifying can comprise performing quantitative PCR, quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RT-PCR), single cell PCR, restriction fragment length polymorphism PCR (PCR-RFLP), PCR-RFLP/RT-PCR-RFLP, hot start PCR, in situ polony PCR, in situ rolling circle amplification (RCA), bridge PCR, picotiter PCR, emulsion PCR, ligase chain reaction (LCR), transcription amplification, self-sustained sequence replication, selective amplification of target polynucleotide sequences, consensus sequence primed polymerase chain reaction (CP-PCR), arbitrarily primed polymerase chain reaction (AP-PCR), degenerate oligonucleotide-primed PCR (DOP-PCR), or nucleic acid sequence based amplification (NASBA).
  • QF-PCR quantitative fluorescent PCR
  • MF-PCR multiplex fluorescent PCR
  • RT-PCR real time PCR
  • the present invention still further provides a method of optimizing cancer therapy for a patient.
  • the method comprises (i) splitting a rare cell-enriched biological sample obtained from the patient at a time point into a plurality of subsamples containing one or more rare cells; (ii) performing a molecular analysis on the one or more rare cells; and (iii) based on the molecular analysis: (a) predicting efficacy of a cancer therapy treatment for the patient; (b) selecting the cancer therapy treatment for the patient; or (c) excluding the cancer therapy treatment for the patient.
  • the molecular analysis can comprise determining the presence or absence of a gene mutation in the one or more rare cells.
  • the rare cell-enriched biological sample can be a rare cell-enriched blood sample.
  • the one or more rare cells can comprise an epithelial cell, a circulating tumor cell, an endothelial cell, or a stem cell.
  • one or more rare cells can comprise an epithelial cell.
  • the rare cell-enriched biological sample can be obtained by rare cell immunoaffinity separation of a biological sample from the patient.
  • the immunoaffinity separation can comprise flowing the biological sample from the patient through an array of obstacles coated with one or more antibodies that selectively bind to rare cells.
  • About one to ten of the rare cells can be contained in at least one of the one or more subsamples.
  • About one to five of the one or more rare cells can be contained in at least one subsample.
  • About one to five of the one or more rare cells can be contained in each of the one or more subsamples.
  • the molecular analysis can further comprise computing a fraction of the plurality of subsamples that contain rare cells having the gene mutation.
  • the patient can have undergone cancer therapy.
  • the cancer therapy can have included administering a composition containing gef ⁇ tinib to the patient.
  • the method further comprises splitting one or more rare cell-enriched biological samples obtained from the patient into a plurality of subsamples at one or more time points subsequent to the time point.
  • the gene mutation can occur in any of the genes listed in Fig. 10.
  • the gene can be EGFR.
  • the gene mutation can occur in any of exons 18-21 of the EGFR gene.
  • the cancer therapy treatment can comprise administering a pharmaceutical composition containing a small molecule inhibitor of EGFR.
  • the molecular analysis can further comprises detecting expression of a gene identified in Figure 10.
  • the gene can be EGFR, EGF, EpCAM, GA733-2, MUC-I, HER-2, or Claudin-7.
  • the gene can be EpCAM.
  • the gene is EGFR or EGF.
  • the level of expression of the gene can be determined, the molecular analysis can comprise performing a molecular beacon assay on the one or more rare cells.
  • Step (i) of the method of optimizing cancer therapy can comprise culturing at least one of the one or more rare cells.
  • the method of optimizing cancer therapy can further comprise clonally expanding the at least one rare cell to obtain a plurality of clonally derived daughter cells.
  • the present invention still further provides a method for selecting a cancer treatment for a patient.
  • the method can comprise performing a molecular analysis on a first daughter cell clonally derived from an isolated rare cell from the patient.
  • the molecular analysis can include detecting the presence or absence of a chemore si stance mutation in the first daughter cell that confers resistance to a first chemotherapeutic agent. If the chemoresistance mutation is detected, a second daughter cell can be subcultured into a plurality of second daughter cell subcultures. At least one of the second daughter cell subcultures can be contacted with an alternative chemotherapeutic agent. At least one second daughter cell subculture can be assayed for sensitivity or resistance to the alternative chemotherapeutic agent.
  • the alternative chemotherapeutic agent can be excluded from the set of candidate chemotherapeutic agents for the cancer treatment.
  • the isolated rare cell can be isolated from a rare cell-enriched biological sample.
  • the rare cell-enriched biological sample can be a rare cell-enriched blood sample.
  • the isolated rare cell can be isolated by splitting the rare cell- enriched blood sample into a plurality of subsamples.
  • the isolated rare cell can be an epithelial cell, a circulating tumor cell, an endothelial cell, or a stem cell.
  • the isolated rare cell can be an epithelial cell.
  • the rare cell-enriched biological sample can be obtained by rare-cell immunoaffinity separation of a biological sample from the patient.
  • the immunoaffinity separation can comprise flowing the biological sample from the patient through an array of obstacles coated with one or more antibodies that selectively bind to rare cells.
  • the molecular analysis can comprise detecting the presence or absence of a mutation in a gene identified in Figure 10.
  • the gene can be an EGFR gene.
  • the mutation can occur in any of exons 18-21 of the EGFR gene.
  • the molecular analysis can comprise detecting expression of a gene identified in Figure 10.
  • the first chemotherapeutic agent can be a small molecule EGFR inhibitor, for example, gefitinib.
  • the plurality of second daughter cell subcultures can be cultured as spheroids.
  • Figures 1A-1E illustrate various embodiments of a size-based separation module.
  • FIGS 2A-2C illustrate one embodiment of an affinity separation module.
  • Figure 3 illustrate one embodiment of a magnetic separation module.
  • Figure 4 illustrates an overview for diagnosing, prognosing, or monitoring a prenatal condition in a fetus.
  • Figure 5 illustrates an overview for diagnosing, prognosing, or monitoring a prenatal condition in a fetus.
  • Figure 6 illustrates an overview for diagnosing, prognosing or monitoring cancer in a patient.
  • Figures 7A-7B illustrate an assay using molecular inversion probes.
  • Figure 7 C illustrates an overview of the use of nucleic acid tags.
  • Figures 8A-8C illustrate one example of a sample splitting apparatus.
  • Figure 9 illustrates the probability of having 2 or more circulating tumor cells loaded into a single sample well.
  • Figure 10 illustrates genes whose expression or mutations can be associated with cancer or another condition diagnosed herein.
  • Figure 11 illustrates primers useful in the methods herein.
  • Figure 12A-B illustrate cell smears of the product and waste fractions.
  • Figure 13A-F illustrate isolated fetal cells confirmed by the reliable presence of male cells.
  • Figure 14 illustrates cells with abnormal trisomy 21 pathology.
  • Figure 15 illustrates performance of a size-based separation module.
  • Figure 16 illustrates histograms of these cell fractions resulting from a size-based separation module.
  • Figure 17 illustrates a first output and a second output of a size-based separation module.
  • Figure 18 illustrates epithelial cells bound to a capture module of an array of obstacles coated with anti-EpCAM.
  • Figures 19A-C illustrate one embodiment of a flow-through size-based separation module adapted to separate epithelial cells from blood and alternative parameters that can be used with such device.
  • Figure 2OA -D illustrate various targeted subpopulations of cells that can be isolated using size- based separation and various cut-off sizes that can be used to separate such targeted subpopulations.
  • Figure 21 illustrates a device of the invention with counting means to determine the number of cells in the enriched sample.
  • Figure 22 illustrates an overview of one aspect of the invention for diagnosing, prognosing, or monitoring cancer in a patient.
  • Figure 23 illustrates the use of EGFR mRNA for generating sequencing templates.
  • Figure 24 illustrates performing real-time quantitative allele-specific PCR reactions to confirm the sequence of mutations in EGFR mRNA.
  • Figure 25 illustrates confirmation of the presence of a mutation is when the signal from a mutant allele probe rises above the background level of fluorescence.
  • Figure 26A-B illustrate the presence of EGFR mRNA in epithelial cells but not leukocytes.
  • Figure 27 illustrate results of the first and second EGFR PCR reactions.
  • Figure 28A-B results of the first and second EGFR PCR reactions.
  • Figure 29 illustrates that EGFR wild type and mutant amplified fragments are readily detected, despite the high leukocyte background.
  • the present invention provides systems, apparatus, and methods to detect the presence of or abnormalities of rare analytes or cells, such as hematopoietic bone marrow progenitor cells, endothelial cells, fetal cells, epithelial cells, or circulating tumor cells (CTCs) in a sample of a mixed analyte or cell population (e.g., maternal peripheral blood samples).
  • rare analytes or cells such as hematopoietic bone marrow progenitor cells, endothelial cells, fetal cells, epithelial cells, or circulating tumor cells (CTCs) in a sample of a mixed analyte or cell population (e.g., maternal peripheral blood samples).
  • Samples containing rare cells can be obtained from any animal in need of a diagnosis or prognosis or from an animal pregnant with a fetus m need of a diagnosis or prognosis
  • a sample can be obtained from an animal suspected of being pregnant, pregnant, or that has been pregnant to detect the presence of a fetus or fetal abnormality.
  • a sample is obtained from an animal suspected of having, having, or an animal that had a disease or condition (e g. cancer) Such a condition can be diagnosed, prognosed, or monitored, and therapy can be determined based on the methods and systems described herein.
  • An animal of the present invention can be a human or a domesticated animal such as a cow, chicken, pig, horse, rabbit, dog, cat, or goat
  • Samples derived from an animal or human can include, e g., whole blood, sweat, tears, ear flow, sputum, lymph, bone marrow suspension, lymph, urine, saliva, semen, vaginal flow, cerebrospinal fluid, brain fluid, ascites, milk, fluid secretions of the respiratory, intestinal, or genitourinary tracts
  • a blood sample can be optionally pre-treated or processed prior to enrichment.
  • pre-treatment steps include the addition of a reagent such as a stabilizer, a preservative, a ftxant, a lysing reagent, a diluent, an anti-apoptotic reagent, a cell viability/inviabihty stain, an anticoagulation reagent, an anti-thrombotic reagent, magnetic property regulating reagent, a buffering reagent, an osmolality regulating reagent, a pH regulating reagent, and/or a cross-linking reagent [0073] When a blood sample is obtained, a preservative such an anti-coagulation agent and/or a stabilizer is often added to the sample prior to enrichment This allows for extended time for analysis/det
  • a sample such as a blood sample
  • a blood sample can be combined with an agent that selectively lyses one or more cells or components in a blood sample
  • fetal cells can be selectively lysed releasing their nuclei when a blood sample including fetal cells is combined with deiomzed water
  • deiomzed water Such selective lysis allows for the subsequent enrichment of fetal nuclei using, e g , size or affinity based separation
  • platelets and/or enucleated red blood cells are selectively lysed to generate a sample enriched m nucleated cells, such as fetal nucleated red blood cells (fnRBCs), maternal nucleated blood cells (mnBC), epithelial cells and CTC
  • the amount can vary depending upon animal size, its gestation period, and the condition being screened In some embodiments, up to 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 mL of a sample is obtained. In some embodiments, 1-50, 2-40, 3- 30, or 4-20 mL of sample is obtained In some embodiments, more than 5, 10, 15, 20, 25, 30, 35, 40, 45,
  • a blood sample can be obtained from a pregnant animal or human within 36, 24, 22, 20, 18, 16, 14, 12, 10, 8, 6 or 4 weeks of gestation.
  • a sample e g , a blood sample
  • can be enriched for rare analytes or rare cells e g fetal cells, epithelial cells or circulating tumor cells
  • rare analytes or rare cells e g fetal cells, epithelial cells or circulating tumor cells
  • the enrichment increases the concentration of rare cells or ratio of rare cells to non-rare cells in the sample.
  • enrichment can increase concentration of an analyte of interest such as a fetal cell or epithelial cell or CTC by a factor of at least 2, 4, 6, 8, 10, 20, 50, 100, 200, 500, 1,000, 2,000, 5,000, 10,000, 20,000, 50,000, 100,000, 200,000, 500,000, 1,000,000, 2,000,000, 5,000,000, 10,000,000, 20,000,000, 50,000,000, 100,000,000, 200,000,000, 500,000,000, 1,000,000,000, 2,000,000,000, or
  • the initial concentration of the fetal cells may be about 1:50,000,000 and it may be increased to at least 1:5,000 or 1:500.
  • Enrichment can also increase concentration of rare cells in volume of rare cells / total volume of sample (removal of fluid).
  • a fluid sample e.g., a blood sample
  • a fluid sample of greater than 10, 15, 20, 50, or 100 mL total volume comprising rare components of interest, and it can be concentrated such that the rare component of interest into a concentrated solution of less than 0.5, 1, 2, 3, 5, or 10 mL total volume.
  • Enrichment can occur using one or more types of separation modules. Several different modules are described herein, all of which can be fiuidly coupled with one another in the series for enhanced performance.
  • enrichment occurs by selective lysis as described above.
  • enrichment of rare cells occurs using one or more size-based separation modules.
  • size-based separation modules include filtration modules, sieves, matrixes, etc.
  • size-based separation modules contemplated by the present invention include those disclosed in International Publication No. WO 2004/113877. Other size based separation modules are disclosed in
  • a size-based separation module comprises one or more arrays of obstacles forming a network of gaps.
  • the obstacles are configured to direct particles as they flow through the array/network of gaps into different directions or outlets based on the particle's hydrodynamic size.
  • nucleated cells or cells having a hydrodynamic size larger than a predetermined certain size such as a cutoff or predetermined size, e.g., 8 ⁇ m
  • a predetermined certain size such as a cutoff or predetermined size, e.g., 8 ⁇ m
  • the enucleated ceils or cells having a hydrodynamic size smaller than a predetermined size, e.g., 8 ⁇ m are directed to a second outlet also located on the opposite side of the array of obstacles from the fluid flow inlet.
  • An array can be configured to separate cells smaller or larger than a predetermined size by adjusting the size of the gaps, obstacles, and offset in the period between each successive row of obstacles.
  • obstacles or gaps between obstacles can be up to 10, 20, 50, 70, 100, 120, 150, 170, or 200 ⁇ m in length or about 2, 4, 6, 8 or 10 ⁇ m in length.
  • an array for size-based separation includes more than 100, 500, 1,000, 5,000, 10,000, 50,000 or 100,000 obstacles that are arranged into more than 10, 20, 50, 100, 200, 500, or 1000 rows.
  • obstacles in a first row of obstacles are offset from a previous (upstream) row of obstacles by up to 50% the period of the previous row of obstacles.
  • obstacles in a first row of obstacles are offset from a previous row of obstacles by up to 45, 40, 35, 30, 25, 20, 15, or 10% the period of the previous row of obstacles.
  • the distance between a first row of obstacles and a second row of obstacles can be up to 10, 20, 50, 70, 100, 120, 150, 170 or 200 ⁇ m.
  • a particular offset can be continuous (repeating for multiple rows) or non-continuous.
  • a separation module includes multiple discrete arrays of obstacles fluidly coupled such that they are in series with one another. Each array of obstacles has a continuous offset, but each subsequent (downstream) array of obstacles has an offset that is different from the previous (upstream) offset. Preferably, each subsequent array of obstacles has a smaller offset that the previous array of obstacles. This allows for a refinement in the separation process as cells migrate through the array of obstacles.
  • a plurality of arrays can be fluidly coupled in series or in parallel,
  • FIG. 1A illustrates an example of a size-based separation module. Obstacles (which may be of any shape) are coupled to a flat substrate to form an array of gaps. A transparent cover or lid may be used to cover the array.
  • the obstacles form a two-dimensional array with each successive row shifted horizontally with respect to the previous row of obstacles, where the array of obstacles directs component having a hydrodynamic size smaller than a predetermined size in a first direction and component having a hydrodynamic size larger that a predetermined size in a second direction.
  • the predetermined size of an array of obstacles can be get at 6-12 ⁇ m or 6-8 ⁇ m.
  • the predetermined size of an array of obstacles can be between 4-10 ⁇ m or 6-8 ⁇ m.
  • the flow of sample into the array of obstacles can be aligned at a small angle (flow angle) with respect to a line-of- sight of the array.
  • the array is coupled to an infusion pump to perfuse the sample through the obstacles.
  • a size-based separation module comprises an array of obstacles configured to direct cells larger than a predetermined size to migrate along a line-of-sight within the array (e.g. towards a first outlet or bypass channel leading to a first outlet), while directing cells and analytes smaller than a predetermined size to migrate through the array of obstacles in a different direction than the larger cells (e.g. towards a second outlet).
  • a line-of-sight within the array
  • directing cells and analytes smaller than a predetermined size to migrate through the array of obstacles in a different direction than the larger cells (e.g. towards a second outlet).
  • a variety of enrichment protocols may be utilized although gentle handling of the cells is needed to reduce any mechanical damage to the cells or their DNA. This gentle handling also preserves the small number of fetal or rare cells in the sample. Integrity of the nucleic acid being evaluated is an important feature to permit the distinction between the genomic material from the fetal or rare cells and other cells in the sample.
  • the enrichment and separation of the fetal or rare cells using the arrays of obstacles produces gentle treatment which minimizes cellular damage and maximizes nucleic acid integrity permitting exceptional levels of separation and the ability to subsequently utilize various formats to very accurately analyze the genome of the cells which are present in the sample in extremely low numbers.
  • enrichment of rare cells e.g.
  • fetal cells, epithelial cells, or circulating tumor cells occurs using one or more capture modules that selectively inhibit the mobility of one or more cells of interest.
  • a capture module is fluidly coupled downstream to a size-based separation module.
  • Capture modules can include a substrate having multiple obstacles that restrict the movement of cells or analytes greater than a predetermined size. Examples of capture modules that inhibit the migration of cells based on size are disclosed in U.S. Patent No. 5,837,115 and 6,692,952.
  • a capture module includes a two dimensional array of obstacles that selectively filters or captures cells or analytes having a hydrodynamic size greater than a particular gap size (predetermined size), International Publication No. WO 2004/113877.
  • a capture module captures analytes (e.g., cells of interest or not of interest) based on their affinity.
  • an affinity-based separation module that can capture cells or analytes can include an array of obstacles adapted for permitting sample flow through, but for the fact that the obstacles are covered with binding moieties that selectively bind one or more analytes (e.g., cell populations) of interest (e.g., red blood cells, fetal cells, epithelial cells, or nucleated cells) or analytes not-of-interest (e.g., white blood cells).
  • Arrays of obstacles adapted for separation by capture can include obstacles having one or more shapes and can be arranged in a uniform or non-uniform order.
  • a two- dimensional array of obstacles is staggered such that each subsequent row of obstacles is offset from the previous row of obstacles to increase the number of interactions between the analytes being sorted (separated) and the obstacles.
  • Binding moieties coupled to the obstacles can include, e.g., proteins (e.g., ligands/receptors), nucleic acids having complementary counterparts in retained analytes, antibodies, etc.
  • an affinity-based separation module comprises a two-dimensional array of obstacles covered with one or more antibodies selected from the group consisting of: anti-CD71, anti-CD235a, anti-CD36, anti-carbohydrates, anti-selectin, anti-CD45, anti-GPA, anti-antigen-i, anti-EpCAM, anti-E-cadherin, and anti-Muc-1.
  • Figure 2A illustrates a path of a first analyte through an array of posts wherein an analyte that does not specifically bind to a post continues to migrate through the array, while an analyte that does bind a post is captured by the array.
  • Figure 2B is a picture of antibody coated posts.
  • Figure 2C illustrates coupling of antibodies to a substrate (e.g., obstacles, side walls, etc.) as contemplated by the present invention. Examples of such affinity-based separation modules are described in International Publication No. WO 2004/029221.
  • a capture module utilizes a magnetic field to separate and/or enrich one or more analytes (cells) based on a magnetic property or magnetic potential in such analyte of interest or an analyte not of interest.
  • analytes cells
  • red blood cells which are slightly diamagnetic (repelled by magnetic field) in physiological conditions can be made paramagnetic (attributed by magnetic field) by deoxygenation of the hemoglobin into methemoglobin. This magnetic property can be achieved through physical or chemical treatment of the red blood cells.
  • a sample containing one or more red blood cells and one or more white blood cells can be enriched for the red blood cells by first inducing a magnetic property in the red blood cells and then separating the red blood cells from the white blood cells by flowing the sample through a magnetic field (uniform or non-uniform).
  • a maternal blood sample can flow first through a size-based separation module to remove enucleated cells and cellular components (e.g., analytes having a hydrodynamic size less than 6 ⁇ ms) based on size.
  • the enriched nucleated cells e.g., analytes having a hydrodynamic size greater than 6 ⁇ ms
  • white blood cells and nucleated red blood cells are treated with a reagent, such as CO 2 , N 2, OrNaNO 2 , that changes the magnetic property of the red blood cells' hemoglobin.
  • a reagent such as CO 2 , N 2, OrNaNO 2
  • the treated sample then flows through a magnetic field (e.g., a column coupled to an external magnet), such that the paramagnetic analytes (e.g., red blood cells) will be captured by the magnetic field while the white blood cells and any other non-red blood cells will flow through the device to result in a sample enriched in nucleated red blood cells (including fetal nucleated red blood cells or fhRBCs).
  • Subsequent enrichment steps can be used to separate the rare cells (e.g. fnRBCs) from the non-rare cells maternal nucleated red blood cells.
  • a sample enriched by size-based separation followed by affinity/magnetic separation is further enriched for rare cells using fluorescence activated cell sorting (FACS) or selective lysis of a subset of the cells.
  • FACS fluorescence activated cell sorting
  • enrichment involves detection and/or isolation of rare cells or rare DNA
  • rare cells e.g. fetal cells or fetal DNA
  • hyperbaric pressure increased levels of CO 2 , e.g., 4% CO 2
  • This will selectively initiate apoptosis in the rare or fragile cells in the sample (e.g , fetal cells).
  • the rare cells e.g. fetal cells
  • their nuclei will condense and optionally be ejected from the rare cells.
  • the rare cells or nuclei can be detected using any technique known in the art to detect condensed nuclei, including DNA gel electrophoresis, in situ labeling of DNA nick using terminal deoxynucleotidyl transferase (TdT)-mediated dUTP in situ nick labeling (TUNEL) (Gavrieli, Y., et al. J. Cell Biol 119:493-501 (1992)), and ligation of DNA strand breaks having one or two-base 3' overhangs (Taq polymerase-based m situ ligation) (Didenko V., et al. J. Cell Biol. 135:1369-76 (1996)).
  • TdT terminal deoxynucleotidyl transferase
  • TUNEL terminal deoxynucleotidyl transferase
  • TUNEL terminal deoxynucleotidyl transferase
  • TUNEL terminal deoxynucleot
  • ejected nuclei can further be detected using a size based separation module adapted to selectively enrich nuclei and other analytes smaller than a predetermined size (e.g. 6 ⁇ ms) and isolate them from cells and analytes having a hydrodynamic diameter larger than 6 ⁇ m.
  • a size based separation module adapted to selectively enrich nuclei and other analytes smaller than a predetermined size (e.g. 6 ⁇ ms) and isolate them from cells and analytes having a hydrodynamic diameter larger than 6 ⁇ m.
  • a predetermined size e.g. 6 ⁇ ms
  • the present invention contemplated detecting fetal cells/fetal DNA and optionally using such fetal DNA to diagnose or prognose a condition m a fetus.
  • Such detection and diagnosis can occur by obtaining a blood sample from the female pregnant with the fetus, enriching the sample for cells and analytes larger than 8 ⁇ m using, for example, an array of obstacles adapted for size-base separation where the predetermined size of the separation is 8 ⁇ m (e.g. the gap between obstacles is up to 8 ⁇ m). Then, the enriched product is further enriched for red blood cells (RBCs) by oxidizing the sample to make the hemoglobin paramagnetic and flowing the sample through one or more magnetic regions. This selectively captures the RBCs and removes other cells (e.g., white blood cells) from the sample.
  • RBCs red blood cells
  • the fnRBCs can be enriched from mnRBCs m the second enriched product by subjecting the second enriched product to hyperbaric pressure or other stimulus that selectively causes the fetal cells to begin apoptosis and condense / eject their nuclei.
  • Such condensed nuclei are then identified/isolated using, e.g , laser capture microdissection or a size based separation module that separates components smaller than 3, 4, 5 or 6 ⁇ m from a sample.
  • Such fetal nuclei can then by analyzed using any method known in the art or described herein.
  • a magnetic particle e g , a bead
  • compound e.g , Fe 3+
  • a bead coupled to an antibody that selectively binds to an analyte of interest can be decorated with an antibody elected from the group of anti CD71 or CD75.
  • a magnetic compound, such as Fe can be couple to an antibody such as those described above.
  • the magnetic particles or magnetic antibodies herein may be coupled to any one or more of the devices herein prior to contact with a sample or may be mixed with the sample prior to delivery of the sample to the device(s). Magnetic particles can also be used to decorate one or more analytes (cells of interest or not of interest) to increase the size prior to performing size-based separation.
  • a magnetic field used to separate analytes/cells in any of the embodiments described herein can be uniform or non-uniform as well as external or internal to the device(s) described herein.
  • An external magnetic field is one whose source is outside a device herein (e.g., container, channel, obstacles).
  • An internal magnetic field is one whose source is withm a device contemplated herein.
  • An example of an internal magnetic field is one where magnetic particles may be attached to obstacles present in the device
  • Analytes captured by a magnetic field can be released by demagnetizing the magnetic regions retaining the magnetic particles.
  • the demagnetization can be limited to selected obstacles or regions.
  • the magnetic field can be designed to be electromagnetic, enabling turn-on and turn-off of the magnetic fields for each individual region or obstacle at will.
  • Figure 3 illustrates an embodiment of a device configured for capture and isolation of cells expressing the transferrin receptor from a complex mixture.
  • Monoclonal antibodies to CD71 receptor are readily available off-the-shelf and can be covalently coupled to magnetic materials comprising any conventional ferroparticles, such as, but not limited to ferrous doped polystyrene and ferroparticles or ferro- colloids (e.g., from Miltenyi and Dynal).
  • the anti CD71 bound to magnetic particles is flowed into the device.
  • the antibody coated particles are drawn to the obstacles (e.g., posts), floor, and walls and are retained by the strength of the magnetic field interaction between the particles and the magnetic field.
  • the particles between the obstacles and those loosely retained with the sphere of influence of the local magnetic fields away from the obstacles are removed by a rmse.
  • One or more of the enrichment modules described herein may be fluidly coupled in series or in parallel with one another.
  • a first outlet from a separation module can be fluidly coupled to a capture module
  • the separation module and capture module are integrated such that a plurality of obstacles acts both to deflect certain analytes according to size and direct them in a path different than the direction of analyte(s) of interest, and also as a capture module to capture, retain, or bind certain analytes based on size, affinity, magnetism or other physical property.
  • the enrichment steps performed have a specificity and/or sensitivity greater than 50, 60, 70, 80, 90, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 99.95%
  • the retention rate of the enrichment module(s) herein is such that >50, 60, 70, 80,
  • analytes or cells of mterest e.g , nucleated cells or red blood cells or nuclei from nucleated cells
  • the enrichment modules are configured to remove >50, 60, 70, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 99.9 % of all unwanted analytes (e.g., red blood-platelet enriched cells) from a sample [00101]
  • Any of the enrichment methods herein may be further supplemented by splitting the enriched sample into ahquots or subsamples.
  • an enriched sample is split into at least 2, 5, 10, 20, 50, 100, 200, 500, or 1000 subsamples.
  • an enriched sample comprises about 500 cells and is split into 500 or 1000 different subsamples, each subsample will have 1 or 0 cells.
  • 5% or more i.e., 10%, 15%, 16%, 17%, 18%, 20%, 25%, 30%, 35%, 50%, 70%, 75%, or any other percent from 5% to 100% of the total number of cells in at least one of the subsamples are rare cells (e.g., epithelial cells, CTCs, or endothelial cells).
  • a sample is split or arranged such that each subsample is in a unique or distinct location (e.g., a well). Such location may be addressable.
  • Each site can further comprise a capture mechanism to capture cell(s) to the site of interest and/or release mechanism for selectively releasing cells from the site of interest.
  • the site is configured to contain a single cell.
  • the methods described herein are used for detecting the presence or conditions of rare cells that are in a mixed sample (optionally even after enrichment) at a concentration of up to 90%, 80%, 70%, 60%, 50 %, 40%, 30%, 20%, 10%, 5% or 1% of all cells in the mixed sample, or at a concentration of less than 1:2, 1:4, 1 :10, 1:50, 1:100, 1:200, 1:500, 1:1000, 1:2000, 1:5000, 1:10,000, 1:20,000, 1:50,000, 1:100,000, 1:200,000, 1:1,000,000, 1:2,000,000, 1.5,000,000, 1:10,000,000, 1:20,000,000, 1:50,000,000 or 1:100,000,000 of all cells in the sample, or at a concentration of less than 1 x
  • the mixed sample has a total of up to 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, or 100 rare cells (e.g., fetal cells or epithelial cells).
  • a peripheral maternal venous blood sample enriched by the methods herein can be analyzed to determine pregnancy or a condition of a fetus (e.g., sex of fetus or trisomy).
  • the analysis step for fetal cells may further include comparing the ratio of maternal to paternal genomic DNA in the identified fetal cells
  • a sample is obtained from an animal, such as a human.
  • the animal or human is pregnant, suspected of being pregnant, or may have been pregnant, and, the systems and methods described herein are used to diagnose pregnancy and/or conditions of the fetus (e.g., trisomy).
  • the animal or human is suspected of having a condition, has a condition, or had a condition (e.g., cancer), and the systems and methods described herein are used to diagnose the condition, determine appropriate therapy, and/or monitor for recurrence.
  • a sample obtained from the animal can be a blood sample, e.g., of up to 50, 40,
  • rare cells e.g., fetal cells or epithelial cells
  • DNA of such rare cells are enriched using one or more methods known m the art or described herein.
  • the sample can be applied to a size-base separation module (e.g., two- dimensional array of obstacles) configured to direct cells or particles in the sample greater than 8 ⁇ m to a first outlet and cells or particles in the sample smaller than 8 ⁇ m to a second outlet.
  • the fetal cells can subsequently be further enriched from maternal white blood cells (which are also greater than 8 ⁇ m) based on their potential magnetic property.
  • N 2 or anti-CD71 coated magnetic beads is added to the first enriched product to make the hemoglobin in the red blood cells (maternal and fetal) paramagnetic.
  • the enriched sample is then flowed through a column coupled to an external magnet.
  • an enriched sample has the rare cells (or rare genomes) consisting of up to 0.01, 0.02, 0.05, 0.1, 0.2, 0.5, 1, 2, 5, 10, 20, or 50% of all cells (or genomes) in the enriched sample.
  • a maternal blood sample of 20 mL from a pregnant human can be enriched for fetal cells such that the enriched sample has a total of about 500 cells, 2% of which are fetal and the rest are maternal.
  • the enriched product is split between two or more discrete locations.
  • a sample is split into at least 2, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000,
  • output from an enrichment module is serially divided into wells of a 1536 microwell plate ( Figure 8). This can result in one cell or genome per location or 0 or 1 cell or genome per location. In some embodiments, cell splitting results in more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 200, 500, 1000, 2000, 5000, 10,000, 20,000, 50,000, 100,000, 200,000, or 500,000 cells or genomes per location. .
  • 5% or more, i.e., 10%, 15%, 16%, 17%, 18%, 20%, 25%, 30%, 35%, 50%, 70%, 75%, or any other percent from 5% to 100% of the total number of genomes in at least one location are rare cell genomes (e.g., genomes from epithelial cells, CTCs, or endothelial cells).
  • the load at each discrete location e.g., well
  • each site includes 0 or 1 fetal cells.
  • Examples of discrete locations which could be used as addressable locations include, but are not limited to, wells, bins, sieves, pores, geometric sites, matrixes, membranes, electric traps, gaps, beads, microspheres, or obstacles.
  • the discrete cells are addressable such that one can correlate a cell or cell sample with a particular location.
  • Examples of methods for splitting a sample into discrete locations include, but are not limited to, fluorescent activated cell sorting (FACS) (Sherlock, JV et al. Ann. Hum. Genet. 62 (Pt. 1): 9-23 (1998)), micromanipulation (Samura, O., Ct al Hum. Genet. 107(l).28-32 (2000)) and dilution strategies (Findlay, I. et al. MoI. Cell. Endocrinol. 183 Suppl 1: S5-12 (2001)).
  • FACS fluorescent activated cell sorting
  • Other methods for sample splitting cell sorting and splitting methods known in the art may also be used.
  • samples can be split by affinity sorting techniques using affinity agents (e.g., antibodies) bound to any immobilized or mobilized substrate (Samura O., et al., Hum Genet. 107(l):28-32 (2000)).
  • affinity agents e.g., antibodies
  • Such affinity agents can be specific to a cell type, e.g., RBCs, fetal cells, epithelial cells, or CTCS, including those that can specifically bind to EpCAM, antigen-i, or CD-71.
  • a sample or enriched sample is transferred to a cell sorting device that includes an array of discrete locations for capturing cells traveling along a fluid flow The discrete locations can be arranged in a defined pattern across a surface such that the discrete sites are also addressable.
  • the sorting device is coupled to any of the enrichment devices known in the art or disclosed herein.
  • Examples of cell sorting devices included are described in International Publication No. WO 01/35071
  • Examples of surfaces that may be used for creating arrays of cells in discrete sites include, but are not limited to, cellulose, cellulose acetate, nitrocellulose, glass, quartz or other crystalline substrates such as gallium arsenide, silicones, metals, semiconductors, various plastics and plastic copolymers, cyclo- olefin polymers, various membranes and gels, microspheres, beads, and paramagnetic or supramagnetic microparticles.
  • a sorting device comprises an array of wells or discrete locations wherein each well or discrete location is configured to hold up to one cell.
  • Each well or discrete location also has a capture mechanism adapted for retention of a cell (e.g., affinity, gravity, suction, etc.) and optionally a release mechanism for selectively releasing a cell of interest from a specific well or site (e.g. bubble actuation).
  • a capture mechanism adapted for retention of a cell
  • a release mechanism for selectively releasing a cell of interest from a specific well or site.
  • the amplified/tagged nucleic acids include at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 90, 90 or 100 polymorphic genomic DNA regions such as short tandem repeats (STRs) or variable number of tandem repeats ("VNTR").
  • STRs short tandem repeats
  • VNTR variable number of tandem repeats
  • STR/s/ are selected for high heterozygosity (variety of alleles) such that the paternal allele of any fetal cell is more likely to be distinct in length from the maternal allele. This results in improved power to detect the presence of fetal cells in a mixed sample and any potential of fetal abnormalities m such cells.
  • STR(s) amplified are selected for their association with a particular condition. For example, to determine fetal abnormality an STR sequence comprising a mutation associated with fetal abnormality or condition is amplified.
  • STRs that can be amplified/analyzed by the methods herein include, but are not limited to D21S1414, D21S1411, D21S1412, D21S11 MBP, D13S634, D13S631, D18S535, AmgXY and XHPRT.
  • Additional STRs that can be amplified/analyzed by the methods herein include, but are not limited to, those at locus F13B (I :q31-q32); TPOX (2:p23-2pter); FIBRA (FGA) (4:q28); CSFIPO (5:q33.3-q34); FI3A (6:p24-p25); THOI (I l :pl5-15.5); VWA (12:pl2-pter); CDU (12pl2-pter); D14S1434
  • STR loci are chosen on a chromosome suspected of trisomy and on a control chromosome. Examples of chromosomes that are often t ⁇ somic include chromosomes 21, 18, 13, and X. In some cases, 1 or more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 STRs are amplified per chromosome tested (Samura, O. et al., CIm. Chem. 47(9): 1622-6 (2001)). For example amplification can be used to generate amplicons of up to
  • PCR primers can include: (i) a primer element, (ii) a sequencing element, and (in) a locator element.
  • the primer element is configured to amplify the genomic DNA region of interest (e.g. STR).
  • the primer element mcludes, when necessary, the upstream and downstream primers for the amplification reactions.
  • Primer elements can be chosen which are multiplexible with other primer pairs from other tags m the same amplification reaction (e.g. fairly uniform melting temperature, absence of cross-priming on the human genome, and absence of p ⁇ mer-pnmer interaction based on sequence analysis)
  • the primer element can have at least 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40 or 50 nucleotide bases, which are designed to specifically hybridize with and amplify the genomic DNA region of interest.
  • the sequencing element can be located on the 5' end of each primer element or nucleic acid tag.
  • the sequencing element is adapted to cloning and/or sequencing of the amplicons.
  • the sequencing element can be about 4, 6, 8, 10, 18, 20, 28, 36, 46 or 50 nucleotide bases in length.
  • the locator which is often incorporated into the middle part of the upstream primer, can include a short DNA or nucleic acid sequence (e.g., about 4, 6, 8, 10, or 20 nucleotide bases). The locator element makes it possible to pool the amplicons from all discrete locations following the amplification step and analyze the amplicons in parallel.
  • Tags are added to the cells/DNA at each discrete location using an amplification reaction.
  • Amplification can be performed using PCR or by a variety of methods including, but not limited to, quantitative PCR, quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RT-PCR), single cell PCR, restriction fragment length polymorphism PCR (PCR-RFLP), PCR- RFLP/RT-PCR-RFLP, hot start PCR, nested PCR, in situ polony PCR, in situ rolling circle amplification (RCA), bridge PCR, picotiter PCR and emulsion PCR.
  • QF-PCR quantitative fluorescent PCR
  • MF-PCR multiplex fluorescent PCR
  • RT-PCR real time PCR
  • PCR-RFLP restriction fragment length polymorphism PCR
  • PCR-RFLP PCR- RFLP/RT-PCR-RFLP
  • hot start PCR nested PCR
  • in situ polony PCR in situ rolling circle amplification
  • RCA in situ rolling circle amplification
  • bridge PCR picotiter PCR
  • Suitable amplification methods include the ligase chain reaction (LCR), transcription amplification, self-sustained sequence replication, selective amplification of target polynucleotide sequences, consensus sequence primed polymerase chain reaction (CP-PCR), arbitrarily primed polymerase chain reaction (AP-PCR), degenerate oligonucleotide-primed PCR (DOP-PCR) and nucleic acid sequence based amplification (NASBA). Additional examples of amplification techniques using PCR primers are described in, U.S. Pat. Nos. 5,242,794, 5,494,810,
  • a further PCR amplification is performed using nested primers for the one or more genomic DNA regions of interest to ensure optimal performance of the multiplex amplification.
  • the nested PCR amplification generates sufficient genomic DNA starting material for further analysis such as in the parallel sequencing procedures below.
  • genomic DNA regions tagged/amplified are pooled and purified prior to further processing. Methods for pooling and purifying genomic DNA are known in the art.
  • pooled genomic DNA/amplicons are analyzed to measure, e.g., allele abundance of genomic DNA regions (e.g. STRs amplified). In some embodiments such analysis involves the use of capillary gel electrophoresis (CGE). In other embodiments, such analysis involves sequencing or ultra deep sequencing.
  • CGE capillary gel electrophoresis
  • Sequencing can be performed using the classic Sanger sequencing method or any other method known in the art.
  • sequencing can occur by sequencing-by-synthesis, which involves inferring the sequence of the template by synthesizing a strand complementary to the target nucleic acid sequence.
  • Sequence-by-synthesis can be initiated using sequencing primers complementary to the sequencing element on the nucleic acid tags.
  • the method involves detecting the identity of each nucleotide immediately after (substantially real-time) or upon (real-time) the incorporation of a labeled nucleotide or nucleotide analog into a growing strand of a complementary nucleic acid sequence in a polymerase reaction. After the successful incorporation of a label nucleotide, a signal is measured and then nulled by methods known in the art. Examples of sequence-by-synthesis methods are described in U.S. Application Publication Nos. 2003/0044781, 2006/0024711, 2006/0024678 and 2005/0100932.
  • labels that can be used to label nucleotide or nucleotide analogs for sequencing-by-synthesis include, but are not limited to, chromophores, fluorescent moieties, enzymes, antigens, heavy metal, magnetic probes, dyes, phosphorescent groups, radioactive materials, chemiluminescent moieties, scattering or fluorescent nanoparticles, Raman signal generating moieties, and electrochemical detection moieties.
  • Sequencing-by- synthesis can generate at least 1,000, at least 5,000, at least 10,000, at least 20,000, 30,000, at least 40,000, at least 50,000, at least 100,000 or at least 500,000 reads per hour. Such reads can have at least 50, at least
  • Another sequencing method involves hybridizing the amplified genomic region of interest to a primer complementary to it. This hybridization complex is incubated with a polymerase, ATP sulfurylase, luciferase, apyrase, and the substrates luciferin and adenosine 5' phosphosulfate. Next, deoxynucleotide triphosphates corresponding to the bases A, C, G, and T (U) are added sequentially. Each base incorporation is accompanied by release of pyrophosphate, converted to ATP by sulfurylase, which drives synthesis of oxyluciferm and the release of visible light. Since pyrophosphate release is equimolar with the number of incorporated bases, the light given off is proportional to the number of nucleotides adding in any one step. The process is repeated until the entire sequence is determined.
  • Yet another sequencing method involves a four-color sequencing by ligation scheme (degenerate ligation), which involves hybridizing an anchor primer to one of four positions. Then an enzymatic ligation reaction of the anchor primer to a population of degenerate nonamers that are labeled with fluorescent dyes is performed. At any given cycle, the population of nonamers that is used is structure such that the identity of one of its positions is correlated with the identity of the fluorophore attached to that nonamer. To the extent that the ligase discriminates for complementa ⁇ ly at that queried position, the fluorescent signal allows the inference of the identity of the base.
  • degenerate ligation involves hybridizing an anchor primer to one of four positions. Then an enzymatic ligation reaction of the anchor primer to a population of degenerate nonamers that are labeled with fluorescent dyes is performed. At any given cycle, the population of nonamers that is used is structure such that the identity of one of its positions is correlated with the identity of the fluoro
  • the anchor primer .nonamer complexes are stripped and a new cycle begins.
  • Methods to image sequence information after performing ligation are known in the art.
  • analysis involves the use of ultra-deep sequencing, such as described in Margmles et al., Nature 437 (7057): 376-80 (2005). Briefly, the amplicons are diluted and mixed with beads such that each bead captures a single molecule of the amplified material. The DNA molecule on each bead is then amplified to generate millions of copies of the sequence which all remain bound to the bead. Such amplification can occur by PCR.
  • Each bead can be placed m a separate well, which can be a (optionally addressable) picohter-sized well.
  • each bead is captured within a droplet of a PCR- reaction-mixture-in-oil-emulsion and PCR amplification occurs withm each droplet.
  • the amplification on the bead results in each bead carrying at least one million, at least 5 million, or at least 10 million copies of the original amplicon coupled to it.
  • the beads are placed into a highly parallel sequencing by synthesis machine which generates over 400,000 reads ( ⁇ 100bp per read) in a single 4 hour run. [00128]
  • Other methods for ultra-deep sequencing that can be used are described in Hong, S. et al. Nat.
  • the role of the ultra-deep sequencing is to provide an accurate and quantitative way to measure the allele abundances for each of the STRs.
  • the total required number of reads for each of the aliquot wells is determined by the number of STRs, the error rates of the multiplex PCR, and the Poisson sampling statistics associated with the sequencing procedures.
  • the enrichment output from step 402 results in approximately 500 cells of which 98% are maternal cells and 2% are fetal cells.
  • Such enriched cells are subsequently split into 500 discrete locations (e.g , wells) in a microtiter plate such that each well contains 1 cell PCR is used to amplify STRs (-3-10 STR loci) on each chromosome of interest.
  • the aneuploidy signal becomes diluted and more loci are needed to average out measurement errors associated with variable DNA amplification efficiencies from locus to locus.
  • the sample division into wells containing ⁇ 1 cell proposed in the methods described herein achieves pure or highly enriched fetal/maternal ratios in some wells, alleviating the requirements for averaging of PCR errors over many loci.
  • T be the fetal/maternal DNA copy ratio in a particular PCR reaction. Trisomy increases the ratio of maternal to paternal alleles by a factor l+f/2. PCR efficiencies vary from allele to allele within a locus by a mean square error in the logarithm given by ⁇ a i le i e 2 , and vary from locus to locus by ⁇ i ocus 2 ; where this second variance is apt to be larger due to differences in primer efficiency.
  • N a is the loci per suspected aneuploid chromosome and N c is the control loci.
  • the squared error expected is the mean of the ln(ratio(m/ ⁇ )), where this mean is taken over N loci is given by 2( ⁇ a n e i e 2 )/N.
  • ⁇ diff 2 2( ⁇ a ,i ele 2 )/N a + 2( ⁇ a!lele 2 )/N c (1)
  • N 144( ⁇ allele /f) 2 (4)
  • the suspected aneuploidy region is usually the entire chromosome and N denotes the number of loci per chromosome.
  • Equation 3 is evaluated for N in the following Table 1 for various values of ⁇ a u e i e and f.
  • sample splitting decreases the number of starting genome copies which increases ⁇ a i le ⁇ e at the same time that it increases the value of fin some wells
  • the methods herein are based on the assumption that the overall effect of splitting is favorable; i.e., that the PCR errors do not increase too fast with decreasing starting number of genome copies to offset the benefit of having some wells with large f.
  • the required number of loci can be somewhat larger because for many loci the paternal allele is not distinct from the maternal alleles, and this incidence depends on the heterozygosity of the loci. In the case of highly polymorphic STRs, this amounts to an approximate doubling of N.
  • the role of the sequencing is to measure the allele abundances output from the amplification step. It is desirable to do this without adding significantly more error due to the Poisson statistics of selecting only a finite number of amplicons for sequencing.
  • the rms error in the ln(abundance) due to Poisson statistics is approximately (N reads ) " 2 - It is desirable to keep this value less than or equal to the PCR error ⁇ a ii e i e -
  • a typical paternal allele needs to be allocated at least ( ⁇ a n e i e ) *2 reads.
  • the mixture input to sequencing contains amplicons from Ni 00 loci of which roughly an abundance fraction f/2 are paternal alleles.
  • the total required number of reads for each of the aliquot wells is given approximately by 2N locl /(f ⁇ a n e i e 2 ).
  • N reads 288 N wells f 3 .
  • step 412 wells with rare cells/alleles (e.g., fetal alleles) are identified.
  • the locator elements of each tag can be used to sort the reads (-200,000 sequence reads) into 'bins' which correspond to the individual wells of the microliter plates ( ⁇ 500 bins).
  • sequence reads from each of the bins are then separated into the different genomic DNA region groups, (e.g. STR loci,) using standard sequence alignment algorithms.
  • the aligned sequences from each of the bins are used to identify rare (e.g., non-maternal) alleles. It is estimated that on average a 15 ml blood sample from a pregnant human will result in -10 bins having a single fetal cell each.
  • an independent blood sample fraction known to contain only maternal cells can be analyzed as described above in order to obtain maternal alleles.
  • This sample can be a white blood cell fraction or simply a dilution of the original sample before enrichment.
  • the sequences or genotypes for all the wells can be similarity-clustered to identify the dominant pattern associated with maternal cells.
  • the detection of non-maternal alleles determines which discrete location (e.g.
  • step 414 condition of rare cells or DNA is determined. This can be accomplished by determining abundance of selected alleles (polymorphic genomic DNA regions) in bin(s) with rare cells/DNA. In some embodiments, allele abundance is used to determine aneuploidy, e.g. chromosomes 13, 18 and 21.
  • Abundance of alleles can be determined by comparing ratio of maternal to paternal alleles for each genomic region amplified (e g., -12 STRs). For example, if 12 STRs are analyzed, for each bin there are 33 sequence reads for each of the STRs. In a normal fetus, a given STR will have 1 1 ratio of the maternal to paternal alleles with approximately 16 sequence reads corresponding to each allele (normal diallelic).
  • the information from the different DNA regions on each chromosome are combined to increase the confidence of a given aneuploidy call.
  • the information from the independent bins containing fetal cells can also be combined to further increase the confidence of the call.
  • the determination of fetal trisomy can be used to diagnose conditions such as, trisomy 13, trisomy 18, trisomy 21 (Down syndrome) and Klinefelter Syndrome (XXY).
  • the methods of the invention allow for the determination of maternal or paternal trisomy.
  • the methods of the invention allow for the determination of trisomy or other conditions in fetal cells in a mixed maternal sample arising from more than one fetus.
  • standard quantitative genotyping technology is used to declare the presence of fetal cells and to determine the copy numbers (ploidies) of the fetal chromosomes.
  • a sample e.g., a mixed sample of rare and non-rare cells
  • the sample is a peripheral maternal blood sample.
  • the sample is enriched for rare cells (e.g., fetal cells) by any method known in the art or described herein. See, e.g., step 402 of Figure 4.
  • step 504 the enriched product is split into multiple distinct sites (e.g., wells). See, e.g., step 404 of Figure 4.
  • PCR primer pairs for amplifying multiple (e.g., 2 - 100) highly polymorphic genomic DNA regions are added to each discrete site or well in the array or microliter plate.
  • highly polymorphic genomic DNA regions e.g., SNPs
  • PCR primer pairs for amplifying SNPs along chromosome 13, 18, 21 and/or X can be designed to detect the most frequent aneuoploidies.
  • Other PCR primer pairs can be designed to amplify SNPs along control regions of the genome where aneuploidy is not expected.
  • the genomic loci e.g., SNPs
  • SNPs in the aneuploidy region or aneuploidy suspect region are selected for high polymorphism such that the paternal alleles of the fetal cells are more likely to be distinct from the maternal alleles. This improves the power to detect the presence of fetal cells in a mixed sample as well as fetal conditions or abnormalities.
  • SNPs can also be selected for their association with a particular condition to be detected in a fetus. In some cases, one or more than one, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 SNPs are analyzed per target chromosome (e.g., 13, 18, 21, and/or X).
  • PCR primers are chosen to be multiplexible with other pairs (fairly uniform melting temperature, absence of cross-priming on the human genome, and absence of primer-primer interaction based on sequence analysis).
  • the primers are designed to generate amplicons 10-200, 20-180, 40-160, 60-140 or 70-100 bp in size to increase the performance of the multiplex PCR.
  • a second of round of PCR using nested primers may be performed to ensure optimal performance of the multiplex amplification.
  • the multiplex amplification of single cells is helpful to generate sufficient starting material for the parallel genotyping procedure.
  • Multiplex PCT can be performed on single cells with minimal levels of allele dropout and preferential amplification. See Sherlock, J., et al. Ann. Hum.
  • amplified polymorphic DNA region(s) of interest e.g , SNPs
  • nucleic acid tags serve two roles: to determine the identity of the different SNPs and to determine the identity of the bin from which the genotype was derived.
  • Nucleic acid tags can comprise primers that allow for allele-specific amplification and/or detection
  • the nucleic acid tags can be of a variety of sizes including up to 10 base pairs, 10-40, 15-30, 18-25 or ⁇ 22 base pair long.
  • a nucleic acid tag comprises a molecular inversion probe (MIP). Examples of MIPs and their uses are described in Hardenbol, P., et al., Nat. Biotechnol. 21(6):673-8 (2003); Hardenbol, P., et al , Genome Res. 15(2):269-75 (2005); and Wang, Y., et al., Nucleic Acids Res. 33(21):el83 (2005).
  • MIP molecular inversion probe
  • FIG 7A illustrates one example of a MIP assay used herein.
  • the MIP tag can include a locator element to determine the identity of the bin from which the genotype was derived. For example, when output from an enrichment procedure results in about 500 cells, the enriched product / cells can be split into a microliter plate containing 500 wells such that each cell is in a different distinct well.
  • Figure 7B illustrates a microliter plate with 500 wells each of which contains a single cell. Each cell is interrogated at 10 different
  • step 510 the tagged amplicons are pooled together for further analysis.
  • the genotype at each polymorphic site is determined and/or quantified using any technique known in the art.
  • genotyping occurs by hybridization of the MIP tags to a microarray containing probes complementary to the sequences of each MIP tag. See US Patent No. 6,858,412.
  • the 20,000 tags are hybridized to a single tag array containing complementary sequences to each of the tagged MIP probes.
  • Microarrays e.g. tag arrays
  • a microarray can have at least 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 500, 1,000, 5,000, 10,000, 15,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, or 100,000 different probes complementary to MIP tagged probes.
  • microarrays capable to monitor several genes according to the methods of the invention are well known m the art.
  • Examples of microarrays that can be used in nucleic acid analysis that may be used are described m US Pat 6,300,063, US Pat 5,837,832, US Pat 6,969, 589, US Pat 6,040,138, US Pat 6,858, 412, US Publication No. 2005/0100893, US Publication No. 2004/0018491, US Publication No. 2003/0215821 and US Publication No. 2003/0207295.
  • step 516 bins with rare alleles (e.g., fetal alleles) are identified.
  • rare allele identification can be accomplished by first using the 22bp tags to sort the 20,000 genotypes into 500 bins which correspond to the individual wells of the original microliter plates. Then, one can identify bins containing non-maternal alleles which correspond to wells that contained fetal cells. Determining the number of bins with non-maternal alleles relative to the total number of bins provides an accurate estimate of the number of fhRBCs that were present in the original enriched cell population When a fetal cell is identified in a given bin, the non-maternal alleles can be detected by 40 independent
  • a condition such as trisomy is determined based on the rare cell polymorphism. For example, after identifying the ⁇ 10 bins that contain fetal cells, one can determine the ploidy of chromosomes 13, 18, 21 and X of such cells by comparing the ratio of maternal to paternal alleles for each of —10 SNPs on each chromosome (X, 13, 18, 21). The ratios for the multiple SNPs on each chromosome can be combined (averaged) to increase the confidence of the aneuploidy call for that chromosome. In addition, the information from the ⁇ 10 independent bins containing fetal cells can also be combined to further increase the confidence of the call.
  • an enriched maternal sample with 500 cells can be split into 500 discrete locations such that each location contains one cell. If ten SNPs are analyzed in each of four different chromosomes, forty tagged MIP probes are added per discrete location to analyze forty different SNPs per cell. The forty SNPs are then amplified in each location using the primer element in the MIP probe as described above. All the amplicons from all the discrete locations are then pooled and analyzed using quantitative genotyping as describe above. In this example a total of 20,000 probes in a microarray are required to genotype the same 40 SNPs in each of the 500 discrete locations (4 chromosomes x 10 SNPs x 500 discrete locations).
  • the above embodiment can also be modified to provide for genotyping by hybridizing the nucleic acid tags to bead arrays as are commercially available by Illumina, Inc. and as described in US Patent Nos. 7,040,959; 7,035,740; 7033,754; 7,025,935, 6,998,274; 6, 942,968; 6,913,884; 6,890,764; 6,890,741;
  • nucleic acid tags An overview of the use of nucleic acid tags is described in Figure 7C.
  • target genomic DNA regions are activated in step 702 such that they may bind paramagnetic particles.
  • assay oligonucleotides, hybridization buffer, and paramagnetic particles are combined with the activated DNA and allowed to hybridize (hybridization step).
  • three oligonucleotides are added for each SNP to be detected. Two of the three oligos are specific for each of the two alleles at a SNP position and are referred to as Allele-Specific Oligos (ASOs).
  • ASOs Allele-Specific Oligos
  • a third oligo hybridizes several bases downstream from the SNP site and is referred to as the Locus-Specific Oligo (LSO). All three oligos contain regions of genomic complementarity (Cl, C2, and C3) and universal PCR primer sites (Pl, P2 and P3). The LSO also contains a unique address sequence (Address) that targets a particular bead type. In some cases, up to 1,536 SNPs may be interrogated in this manner.
  • the assay oligonucleotides hybridize to the genomic DNA sample bound to paramagnetic particles. Because hybridization occurs prior to any amplification steps, no amplification bias is introduced into the assay.
  • the above primers can further be modified to serve the two roles of determining the identity of the different SNPs and to determining the identity of the bin from which the genotype was derived.
  • step 704 following the hybridization step, several wash steps are performed reducing noise by removing excess and mis-hybridized oligonucleotides.
  • Extension of the appropriate ASO and ligation of the extended product to the LSO joins information about the genotype present at the SNP site to the address sequence on the LSO.
  • the joined, full-length products provide a template for performing PCR reactions using universal PCR primers Pl, P2, and P3. Universal primers Pl and P2 are labeled with two different labels (e.g., Cy3 and Cy5).
  • labels that can be used include, chromophores, fluorescent moieties, enzymes, antigens, heavy metal, magnetic probes, dyes, phosphorescent groups, radioactive materials, chemiluminescent moieties, scattering or fluorescent nanoparticles, Raman signal generating moieties, or electrochemical detection moieties.
  • step 706 the single-stranded, labeled DNAs are eluted and prepared for hybridization.
  • step 707 the single-stranded, labeled DNAs are hybridized to their complement bead type through their unique address sequence. Hybridization of the GoldenGate Assay products onto the Array Matrix of Beadchip allows for separation of the assay products in solution, onto a solid surface for individual SNP genotype readout.
  • step 708 the array is washed and dried.
  • a reader such as the BeadArray Reader is used to analyze signals from the label.
  • the labels are dye labels such as Cy3 and Cy5
  • the reader can analyze the fluorescence signal on the Sentrix Array Matrix or BeadChip.
  • a computer readable medium having a computer executable logic recorded on it can be used in a computer to perform receive data from one or more quantified DNA genomic regions to automate genotyping clusters and callings. Expression detection and analysis using microarrays is described in part in VaIk, P. J. et al. New England Journal of Medicine 350(16), 1617-28, 2004; Modlich, O. et al.
  • any of the embodiments described herein preferably, more than 1000, 5,000, 10,000, 50,000, 100,000, 500,000, or 1,000,000 SNPs are interrogated in parallel.
  • the systems and methods herein can be used to diagnose, prognose, and monitor neoplastic conditions such as cancer in a patient.
  • neoplastic conditions contemplated herein include acute lymphoblastic leukemia, acute or chronic lymphocyctic or granulocytic tumor, acute myeloid leukemia, acute promyelocytic leukemia, adenocarcinoma, adenoma, adrenal cancer, basal cell carcinoma, bone cancer, brain cancer, breast cancer, bronchi cancer, cervical dysplasia, chronic myelogenous leukemia, colon cancer, epidermoid carcinoma,
  • Cancers such as breast, colon, liver, ovary, prostate, and lung as well as other tumors exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells, e.g., epithelial cells into the bloodstream.
  • epithelial cells exfoliate cells,
  • a biological sample is obtained from an animal such as a human.
  • the human can be suspected of having cancer or cancer recurrence or may have cancer and is in need of therapy selection.
  • the biological sample obtained is a mixed sample comprising normal cells as well as one or more CTCs, epithelial cells, endothelial cells, stem cells, or other cells indicative of cancer.
  • the biological sample is a blood sample.
  • multiple biological samples are obtained from the animal at different points in time (e.g., regular intervals such as daily, or every 2, 3 or 4 days, weekly, bimonthly, monthly, bi-yearly or yearly.
  • step 602 the mixed sample is then enriched for epithelial cells or CTCs or other cells indicative of cancer.
  • Epithelial cells that are exfoliated from solid tumors have been found in very low concentrations in the circulation of patients with advanced cancers of the breast, colon, liver, ovary, prostate, and lung, and the presence or relative number of these cells in blood has been correlated with overall prognosis and response to therapy.
  • These epithelial cells which are in fact CTCs, can be used as an early indicator of tumor expansion or metastasis before the appearance of clinical symptoms.
  • CTCs are generally larger than most blood cells.
  • one useful approach for isolating CTCs from blood is to enrich the biological sample for them based on size, resulting in a cell population enriched in CTCs.
  • Another way to enrich CTCs is by affinity separation, using antibodies specific for particular cell surface markers may be used.
  • Useful endothelial cell surface markers include CD 105, CD 106, CD 144, and CD146; useful tumor endothelial cell surface markers include TEMl, TEM5, and TEM8 (see, e.g., Carson- Walter et al., Cancer Res. 61:6649-6655 (2001)); and useful mesenchymal cell surface markers include
  • CD 133 Antibodies to these or other markers may be obtained from, e.g., Chemicon, Abeam, and R&D Systems.
  • a size-based separation module that enriches CTCs from a fluid sample (e.g., blood) comprises an array of obstacles that selectively deflect particles having a hydrodynamic size larger than 10 ⁇ m into a first outlet and particles having a hydrodynamic size smaller than 10 ⁇ m into a second outlet is used to enrich epithelial cells and CTCs from the sample.
  • step 603 the enriched product is split into a plurality of discrete sites, such as micro wells.
  • Exemplary microwells that can be used in the present invention include microplates having 1536 wells as well as those of lesser density (e.g., 96 and 384 wells).
  • Microwell plate designs contemplated herein include those have 14 outputs that can be automatically dispensed at the same time, as well as those with
  • Figure 9 illustrates one embodiments of a microwell plate contemplated herein.
  • dispensing of the cells into the various discrete sites is automated. In some cases, about 1, 5, 10, or 15 ⁇ L of enriched sample is dispensed into each well. Preferably, the size of the well and volume dispensed into each well is such that only 1 cell is dispensed per well and only 1-5 or less than 3 cells can fit in each well.
  • An exemplary array for sample splitting is illustrated in Figure 8A.
  • Figure 8B illustrates an isometric view and Figure 8B illustrates a top view and cross sectional view of such an array. A square array of wells is arranged such that each subsequent row or column of wells is identical to the previous row or column of wells, respectively.
  • an array of wells is configured in a substrate or plate that about 2.0 cm 2 , 2.5 cm 2 , 3 cm 2 or larger.
  • the wells can be of any shape, e.g., round, square, or oval.
  • the height or width of each well can be between 5-50 ⁇ m, 10-40 ⁇ m, or about 25 ⁇ m.
  • the depth of each well can be up to 100, 80, 60, or 40 ⁇ m; and the radius between the centers of two wells in one column is between 10-60 ⁇ m, 20-50 ⁇ m, or about 35 ⁇ m.
  • an array of wells of area 2.5 cm 2 can have a at least 0.1 x 10 6 wells, 0.2 x 10 6 wells, 0.3 x 10 6 wells, 0.4 x 10 6 wells, or 0.5 x 10 6 wells.
  • each well may have an opening at the bottom.
  • the bottom opening is preferably smaller in size than the cells ⁇ of interest.
  • the bottom opening of each well can have a radius of up to 8, 7, 6, 5, 4, 3, 2 or 1 ⁇ m.
  • the bottom opening allows for cells non-of interest and other components smaller than the cell of interest to be removed from the well using flow pressure, leaving the cells of interest behind in the well for further processing.
  • the array of wells can be a micro-electro-mechanical system (MEMS) such that it integrates mechanical elements, sensors, actuators, and electronics on a common silicon substrate through microfabrication technology.
  • MEMS micro-electro-mechanical system
  • Any electronics in the system can be fabricated using integrated circuit (IC) process sequences (e.g., CMOS, Bipolar, or BICMOS processes), while the micromechanical components are fabricated using compatible micromachining processes that selectively etch away parts of the silicon wafer or add new structural layers to form the mechanical and electromechanical devices.
  • IC integrated circuit
  • CMOS complementary metal-electro-mechanical system
  • the array of wells can be coupled to a microscope slide or other substrate that allows for convenient and rapid optical scanning of all chambers (i.e. discrete sites) under a microscope.
  • a 1536-well microliter plate is used for enhanced convenience of reagent addition and other manipulations.
  • the enriched product can be split into wells such that each well is loaded with a plurality of leukocytes (e.g., more than 100, 200, 500, 1000, 2000, or 5000).
  • leukocytes are dispensed per well, while random wells will have a single rare cell or up to 2, 3, 4, or 5 rare cells (e.g, epithelial cells, CTCs, or endothelial cells).
  • rare cells e.g, epithelial cells, CTCs, or endothelial cells.
  • 5% or more, i.e., 10%, 15%, 16%, 17%, 18%, 20%, 25%, 30%, 35%, 50%, 70%, 75%, or any other percent from 5% to 100% of the total number of cells in at least one of the wells are rare cells.
  • the probability of getting a single epithelial cell or CTC into a well is calculated such that no more than 1 CTC is loaded per well.
  • the probability of dispensing CTCs from a sample into wells can be calculated using Poisson statistics. When dispensing a 15 mL sample into 1536 well plate at 10 ⁇ L per well, it is not until the number of CTCs in the sample is > 100 that there is more than negligible probability of two or more CTCs being loaded into the sample well.
  • Figure 9 illustrates the probability density function of loading two CTCs into the same plate.
  • step 604 rare cells (e.g., epithelial cells or CTCs) or rare DNA is detected and/or analyzed in each well.
  • detection and analysis includes enumerating epithelial cells and/or CTCs.
  • step 604 involves enumerating CTC and/or epithelial cells in a sample (array of wells) and determining based on their number if a patient has cancer, severity of condition, therapy to be used, or effectiveness of therapy administered.
  • the method herein involve making a series of measurements, optionally made at regular intervals such as one day, two days, three days, one week, two weeks, one month, two months, three months, six months, or one year, or any other interval between one day and one year, one may track the level of epithelial cells present in a patient's bloodstream as a function of time.
  • this provides a useful indication of the progression of the disease and assists medical practitioners in making appropriate therapeutic choices based on the increase, decrease, or lack of change in epithelial cells, e.g., CTCs, in the patient's bloodstream.
  • a sudden increase in the number of cells detected may provide an early warning that the patient has developed a tumor. This early diagnosis, coupled with subsequent therapeutic intervention, is likely to result in an improved patient outcome in comparison to an absence of diagnostic information.
  • more than one type of cell e.g., epithelial, endothelial, etc.
  • a determination of a ratio of numbers of cells or profile of various cells can be obtained to generate the diagnosis or prognosis.
  • the fraction of subsamples that contain one or more rare cells is determined, without necessarily enumerating the number of rare cells in each subsample.
  • detection of rare cells or rare DNA can be made by detecting one or more cancer biomarkers, e.g., any of those listed in Figure 10 in one or more cells in the array. Detection of cancer biomarkers can be accomplished using, e.g., an antibody specific to the marker or by detecting a nucleic acid encoding a cancer biomarker, e.g., listed in Figure 9.
  • single cell analysis techniques are used to analyze individual cells in each well.
  • single cell PCR may be performed on a single cell in a discrete location to detect one or more mutant alleles in the cell (Thornhill AR, J. MoI. Diag; (4) 11-29 (2002)) or a mutation in a gene listed in Figure 9.
  • gene expression analysis can be performed even when the number of cells per well is very low (e.g., one cell per well) using techniques known in the art. (Giordano et al., Am. J. Pathol.
  • single cell expression analysis can be performed to detection expression of one or more genes of interest (Liss B., Nucleic Acids Res., 30 (2002)) including those listed in Figure 9.
  • ultra-deep sequencing can be performed on single cells using methods such as those described in Marguiles M., et al. Nature, "Genome sequencing in microfabricated high-density picolitre reactors.” DOI 10.1038, in which whole genomes are fragmented, fragments are captured using common adapters on their own beads and within droplets of an emulsion, clonally amplified.
  • Such ultra-deep sequencing can also be used to detect mutations in genes associated with cancer, such as those listed in Figure 9.
  • fluorescence in- situ hybridization can be used, e.g., to determine the tissue or tissues of origin of the cells being analyzed.
  • morphological analyses are performed on the cells in each well. Morphological analyses include identification, quantification and characterization of mitochondrial DNA, telomerase, or nuclear matrix proteins. Parrella et al., Cancer Res. 61:7623-7626 (2001); Jones et al., Cancer Res. 61:1299-1304 (2001); Fliss et al., Science 287:2017-2019 (2000); and Soria et al., Clin. Cancer Res. 5:971-
  • the molecular analyses involves determining whether any mitochrondial abnormalities or whether perinuclear compartments are present. Carew et al., MoI. Cancer 1:9 (2002); and Wallace, Science 283:1482-1488 (1999). [00179] A variety of cellular characteristics may be measured using any technique known in the art, including: protein phosphorylation, protein glycosylation, DNA methylation (Das et al., J. Clin. Oncol.
  • microRNA levels He et al., Nature 435:828-833 (2005), Lu et al., Nature 435:834- 838 (2005), O'Donnell et al., Nature 435:839-843 (2005), and Calin et al., N. Engl. J. Med. 353:1793-1801 (2005)
  • cell morphology or other structural characteristics e.g., pleomorphisms, adhesion, migration, binding, division, level of gene expression, and presence of a somatic mutation.
  • This analysis may be performed on any number of cells, including a single cell of interest, e.g., a cancer cell.
  • the cell(s) in each well are lysed and RNA is extracted using any means known in the art.
  • the Quiagen RNeasyTM 96 bioRobotTM 8000 system can be used to automate high-throughput isolation of total RNA from each discrete site.
  • reverse transcriptase reactions can be performed to generate cDNA, which can then be used for performing multiplex PCR reactions on target genes.
  • target genes For example, 1 or more than 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 target genes can be amplified in the same reaction.
  • primers are chosen to be multiplexable (fairly uniform melting temperature, absence of cross -priming on the human genome, and absence of primer-primer interaction based on sequence analysis) with other pairs of primers.
  • Multiple dyes and multi-color fluorescence readout may be used to increase the multiplexing capacity.
  • dyes that can be used to label primers for amplification include, but are not limited to, chromophores, fluorescent moieties, enzymes, antigens, heavy metal, magnetic probes, dyes, phosphorescent groups, radioactive materials, chemilumine scent moieties, scattering or fluorescent nanoparticles, Raman signal generating moieties, and electrochemical detection moieties.
  • PCR amplification can be performed on genes that are expressed in epithelial cells and not in normal cells, e.g., white blood cells or other cells remaining in an enriched product.
  • genes that can be analyzed according to the methods herein include EGFR, EpCAM, GA733-2, MUC-I, HER-2, Claudin-7 and any other gene identified in Figure 10.
  • analysis of the expression level or pattern of such a polypeptide or nucleic acid e.g., cell surface markers, genomic DNA, mRNA, or microRNA, may result in a diagnosis or prognosis of cancer.
  • analysis step 604 involves identifying cells from a mixed sample that express genes which are not expressed in the non-rare cells (e.g. EGFR or EpCAM).
  • a mixed sample that express genes which are not expressed in the non-rare cells (e.g. EGFR or EpCAM).
  • an important indicator for circulating tumor cells is the presence/expression of EGFR or EGF at high levels wherein non-cancerous epithelial cells will express EGFR or EGF at smaller amounts if at all.
  • the presence or absence of certain mutations in EGFR can be associated with diagnosis and/or prognosis of the cancer as well and can also be used to select a more effective treatment (see, e.g., International Publication WO 2005/094357).
  • EGFR inhibitors such as gefitinib (Iressa; AstraZeneca)
  • Exemplary mutations that can be analyzed include those clustered around the ATP-brnding pocket of the EGFR tyrosine kinase (TK) domain, which are known to make cells susceptible to gefitinib inhibition
  • TK tyrosine kinase
  • the patient can be diagnosed with cancer or lack thereof
  • the patient can be prognosed with a particular type of cancer
  • therapy may be determined based on the types of mutations detected
  • cancer cells may be detected in a mixed sample (e g CTCs and circulating normal cells) using one or more of the sequencing methods described herein Briefly, RNA is extracted from cells in each location and converted to cDNA as described above Target genes are then amplified and high throughput ultra deep sequencing is performed to detect a mutation expression level associated with cancer [00189]
  • a mutated gene mRNA e g , mRNA from a mutated EGFR gene
  • rare cells e g , epithelial cells
  • rare cells are cultured (e g , in single cell cultures)
  • cultured rare cells are tested with one or molecular beacon probes to detect mutated gene mRNAs as described above
  • individual cultured rare cells that test positive for the presence of a mutated gene mRNA (e g., a mutated EGFR mRNA) m a molecular beacon assay can be passaged to yield clonally derived daughter cells
  • the daughter cells can subsequently be passaged and/or expanded as needed m a microwell format as described in, e g , Rettig et al , Anal Chem Sep 1,77(17) 5628-5634 (2005)
  • all cultured rare cells are clonally expanded and passaged
  • the passaged clonal daughter cells can then used for genetic analysis as described herein and/or responsiveness to one or more cancer treatments
  • genetic analysis is performed at an early passage (e g , 5 or fewer passages
  • genetic analysis is used to identify one or more rare cell clones bearing one or more mutations (e.g., an EGFR mutation) associated with resistance to a chemotherapeutic agent ("chemoresistance mutations").
  • mutant clones Individual rare cell clones (“mutant clones") identified by any of the methods described herein as bearing the mutations can then be expanded and tested in vitro for sensitivity to a battery of cancer treatments including, but not limited to, chemotherapeutic agents, combinations of chemotherapeutic agents, chemosensitizer agents, radiation therapies, radiosensitizer agents, photodynamic therapies, and photothermal therapies. Cancer treatment modalities identified as particularly effective against mutant clones are then selected for use on a patient from which the rare cell clones were derived.
  • cancer treatment can be optimized for an individual patient by testing a wide range of cancer therapy treatments on the types of cells from the patient that are likely to be refractory to many cancer therapies, i.e., cancer cells bearing chemoresistance mutations.
  • a follow-up analysis can be performed to identify new mutations or changes in the frequencies of mutations in rare cells (e.g., CTCs) isolated from the treated patient.
  • any of the steps herein can be performed using computer program product that comprises a computer executable logic recorded on a computer readable medium.
  • the computer program can use data from target genomic DNA regions to determine the presence or absence of fetal cells in a sample and to determine fetal abnormalities in detected cells.
  • computer executable logic uses data input on STR or SNP intensities to determine the presence of fetal cells in a test sample and determine fetal abnormalities and/or conditions in said cells.
  • the computer program may be specially designed and configured to support and execute some or all of the functions for determining the presence of rare cells such as fetal cells or epithelial/CTCs in a mixed sample and abnormalities and/or conditions associated with such rare cells or their DNA including the acts of (i) controlling the splitting or sorting of cells or DNA into discrete locations (n) amplifying one or more regions of genomic DNA e.g. trisomic region(s) and non-t ⁇ somic region(s) (particularly DNA polymorphisms such as STR and SNP) in cells from a mixed sample and optionally control sample,
  • the program can fit data of the quantity of allele abundance for each polymorphism into one or more data models.
  • One example of a data model provides for a determination of the presence or absence of aneuploidy using data of amplified polymorphisms present at loci in DNA from samples that are highly enriched for fetal cells. The determination of presence of fetal cells m the mixed sample and fetal abnormalities and/or conditions in said cells can be made by the computer program or by a user [00194] In one example, let T be the fetal/maternal DNA copy ratio in a particular PCR reaction.
  • Trisomy increases the ratio of maternal to paternal alleles by a factor l+f/2.
  • PCR efficiencies vary from allele to allele within a locus by a mean square error in the logarithm given by ⁇ a u e ] e 2 , and vary from locus to locus by ⁇ i ocus 2 , where this second variance is apt to be larger due to differences in primer efficiency.
  • N a is the loci per suspected aneuploid chromosome and N c is the control loci.
  • the squared error expected is the mean of the ln(ratio(m/p)), where this mean is taken over N loci is given by 2( ⁇ a u e i e 2 )/N.
  • N 144( ⁇ allele /f) 2 (4)
  • the suspected aneuploidy region is usually the entire chromosome and N denotes the number of loci per chromosome.
  • Equation 3 is evaluated for N in Table 1 for various values of ⁇ a ⁇ e i e and f.
  • the role of the sequencing is to measure the allele abundances output from the amplification step.
  • the rms error in the ln(abundance) due to Poisson statistics is approximately (N readS ) ⁇ i/2 - It is desirable to keep this value less than or equal to the PCR error ⁇ a ii e i e -
  • a typical paternal allele needs to be allocated at least ( ⁇ al i e i e ) "2 reads.
  • the mixture input to sequencing contains amplicons from N locl loci of which roughly an abundance fraction f/2 are paternal alleles.
  • the total required number of reads for each of the aliquot wells is given approximately by 2N ]0C1 /(f ⁇ a u e i e 2 ).
  • N reads 288 N we ii s f 3 .
  • the program can determine the total number of reads that need to be obtained for determining the presence or absence of aneuploidy in a patient sample.
  • the computer program can work in any computer that may be any of a variety of types of general- purpose computers such as a personal computer, network server, workstation, or other computer platform now or later developed.
  • a computer program product is described comprising a computer usable medium having the computer executable logic (computer software program, including program code) stored therein.
  • the computer executable logic can be executed by a processor, causing the processor to perform functions described herein.
  • some functions are implemented primarily in hardware using, for example, a hardware state machine. Implementation of the hardware state machine so as to perform the functions described herein will be apparent to those skilled in the relevant arts.
  • the computer executing the computer logic of the invention may also include a digital input device such as a scanner.
  • the digital input device can provide an image of the target genomic DNA regions (e.g. DNA polymorphism, preferably STRs or SNPs) according to method of the invention.
  • the scanner can provide an image by detecting fluorescent, radioactive, or other emissions; by detecting transmitted, reflected, or scattered radiation; by detecting electromagnetic properties or characteristics; or by other techniques. Various detection schemes are employed depending on the type of emissions and other factors.
  • the data typically are stored in a memory device, such as the system memory described above, in the form of a data file.
  • the scanner may identify one or more labeled targets.
  • a first DNA polymorphism may be labeled with a first dye that fluoresces at a particular characteristic frequency, or narrow band of frequencies, in response to an excitation source of a particular frequency.
  • a second DNA polymorphisms may be labeled with a second dye that fluoresces at a different characteristic frequency.
  • the excitation sources for the second dye may, but need not, have a different excitation frequency than the source that excites the first dye, e.g., the excitation sources could be the same, or different, lasers.
  • a human being may inspect a printed or displayed image constructed from the data in an image file and may identify the data (e.g. fluorescence from microarray) that are suitable for analysis according to the method of the invention.
  • the information is provided in an automated, quantifiable, and repeatable way that is compatible with various image processing and/or analysis techniques.
  • kits which permit the enrichment and analysis of the rare cells present in small qualities in the samples.
  • kits may include any materials or combination of materials described for the individual steps or the combination of steps ranging from the enrichment through the genetic analysis of the genomic material.
  • the kits may include the arrays used for size-based separation or enrichment, labels for uniquely labeling each cell, the devices utilized for splitting the cells into individual addressable locations and the reagents for the genetic analysis.
  • a kit might contain the arrays for size-based separation, unique labels for the cells and reagents for detecting polymorphisms including STRs or SNPs, such as reagents for performing PCR.
  • Figure IE shows a schematic of the device used to separate nucleated cells from fetal cord blood.
  • Dimensions 100 mm * 28 mm x 1mm
  • Device fabrication The arrays and channels were fabricated in silicon using standard photolithography and deep silicon reactive etching techniques. The etch depth is 140 ⁇ m. Through holes for fluid access are made using KOH wet etching. The silicon substrate was sealed on the etched face to form enclosed fluidic channels using a blood compatible pressure sensitive adhesive (9795, 3M, St Paul, MN).
  • Device packaging The device was mechanically mated to a plastic manifold with external fluidic reservoirs to deliver blood and buffer to the device and extract the generated fractions.
  • Device operation An external pressure source was used to apply a pressure of 2.0 PSI to the buffer and blood reservoirs to modulate fluidic delivery and extraction from the packaged device.
  • Experimental conditions Human fetal cord blood was drawn into phosphate buffered saline containing Acid Citrate Dextrose anticoagulants. ImL of blood was processed at 3 mL/hr using the device described above at room temperature and within 48 hrs of draw.
  • Nucleated cells from the blood were separated from enucleated cells (red blood cells and platelets), and plasma delivered into a buffer stream of calcium and magnesium-free Dulbecco's Phosphate Buffered Saline (14190-144, Invitrogen, Carlsbad, CA) containing 1% Bovine Serum Albumin (BSA) (A8412- 100ML, Sigma-Aldrich, St Louis, MO) and 2 mM
  • BSA Bovine Serum Albumin
  • Example 1 The device and process described in detail in Example 1 were used in combination with immunomagnetic affinity enrichment techniques to demonstrate the feasibility of isolating fetal cells from maternal blood.
  • Experimental conditions blood from consenting maternal donors carrying male fetuses was collected into K 2 EDTA vacutainers (366643, Becton Dickinson, Franklin Lakes, NJ) immediately following elective termination of pregnancy. The undiluted blood was processed using the device described in Example 1 at room temperature and within 9 hrs of draw.
  • Nucleated cells from the blood were separated from enucleated cells (red blood cells and platelets), and plasma delivered into a buffer stream of calcium and magnesium-free Dulbecco's Phosphate Buffered Saline (14190-144, Invitrogen, Carlsbad, CA) containing 1% Bovine Serum Albumin (BSA) (A8412-100ML, Sigma-Aldrich, St Louis, MO).
  • BSA Bovine Serum Albumin
  • the nucleated cell fraction was labeled with anti-CD71 microbeads (130-046-201, Miltenyi Biotech Inc., Auburn, CA) and enriched using the MiniMACSTM MS column (130-042-201, Miltenyi Biotech Inc., Auburn, CA) according to the manufacturer's specifications. Finally, the CD 71 -positive fraction was spotted onto glass slides.
  • Measurement techniques Spotted slides were stained using fluorescence in situ hybridization (FISH) techniques according to the manufacturer's specifications using Vysis probes (Abbott Laboratories, Downer's Grove, IL). Samples were stained from the presence of X and Y chromosomes. In one case, a sample prepared from a known Trisomy 21 pregnancy was also stained for chromosome 21. [00217] Performance: Isolation of fetal cells was confirmed by the reliable presence of male cells in the
  • Fetal cells or nuclei can be isolated as described in the enrichment section or as described in example 1 and example 2. Quantitative genotyping can then be used to detect chromosome copy number changes.
  • Figure 5 depicts a flow chart depicting the major steps involved in detecting chromosome copy number changes using the methods described herein.
  • the enrichment process described in example 1 may generate a final mixture containing approximately 500 maternal white blood cells (WBCs), approximately 100 [maternal nuclear red blood cells] (mnBCs), and a minimum of approximately 10 fetal nucleated red blood cells (fhRBCs) starting from an initial 20 ml blood sample taken late in the first trimester.
  • WBCs maternal white blood cells
  • mnBCs approximately 100 [maternal nuclear red blood cells]
  • fhRBCs fetal nucleated red blood cells
  • PCR primer pairs for multiple (40 — 100) highly polymorphic SNPs can then be added to each well in the microtiter plate.
  • SNPs primers can be designed along chromosomes 13, 18, 21 and X to detect the most frequent aneuploidies, and along control regions of the genome where aneuploidy is not expected.
  • Multiple ( ⁇ 10) SNPs would be designed for each chromosome of interest to allow for non- informative genotypes and to ensure accurate results.
  • PCR primers would be chosen to be multiplexible with other pairs (fairly uniform melting temperature, absence of cross-priming on the human genome, and absence of primer-primer interaction based on sequence analysis).
  • the primers would be designed to generate amplicons 70 - 100 bp in size to increase the performance of the multiplex PCR.
  • the primers would contain a 22 bp tag on the 5' which is used in the genotyping analysis.
  • a second of round of PCR using nested primers may be performed to ensure optimal performance of the multiplex amplification.
  • the Molecular Inversion Probe (MIP) technology developed by Affymetrix (Santa Clara, CA) can genotype 20,000 SNPs or more in a single reaction. In the typical MIP assay, each SNP would be assigned a 22bp DNA tag which allows the SNP to be uniquely identified during the highly parallel genotyping assay.
  • the DNA tags serve two roles: (1) determine the identity of the different SNPs and (2) determine the identity of the well from which the genotype was derived. For example, a total of 20,000 tags would be required to genotype the same 40 SNPs in 500 wells different wells (4 chromosomes x 10 SNPs x 500 wells)
  • the tagged MIP probes would be combined with the amplicons from the initial multiplex single- cell PCR and the genotyping reactions would be performed.
  • the probe/template mix would be divided into 4 tubes each containing a different nucleotide (e.g. G, A, T or C).
  • a different nucleotide e.g. G, A, T or C.
  • the mixture would be treated with exonuclease to remove all linear molecules and the tags of the surviving circular molecules would be amplified using PCR.
  • the amplified tags form all of the bins would then be pooled and hybridized to a single DNA microarray containing the complementary sequences to each of the 20,000 tags.
  • Identify bins with non-maternal alleles i.e., fetal cells:
  • the first step in the data analysis procedure would be to use the 22 bp tags to sort the 20,000 genotypes into bins which correspond to the individual wells of the original microtiter plates.
  • the second step would be to identify bins contain non- maternal alleles which correspond to wells that contained fetal cells. Determining the number bins with non-maternal alleles relative to the total number of bins would provide an accurate estimate of the number of fnRBCs that were present in the original enriched cell population.
  • the non-maternal alleles would be detected by 40 independent SNPs which provide an extremely high level of confidence in the result.
  • Detect ploidy for chromosomes 13, 18, and 21 After identifying approximately 10 bins that contain fetal cells, the next step would be to determine the ploidy of chromosomes 13, 18, 21 and X by comparing ratio of maternal to paternal alleles for each of the 10 SNPs on each chromosome. The ratios for the multiple SNPs on each chromosome can be combined (averaged) to increase the confidence of the aneuploidy call for that chromosome. In addition, the information from the approximate 10 independent bins containing fetal cells can also be combined to further increase the confidence of the call.
  • Example 4 Ultra-deep Sequencing for Trisomy Diaenosis on Fetal Cells
  • Fetal cells or nuclei can be isolated as described in the enrichment section or as described in example 1 and example 2. Ultra deep sequencing methods can then be used to detect chromosome copy number changes.
  • Figure 4 depicts a flow chart depicting the major steps involved in detecting chromosome copy number changes using the methods described herein.
  • the enrichment process described in example 1 may generate a final mixture containing approximately 500 maternal white blood cells (WBCs), approximately 100 maternal nuclear red blood cells (mnBCs), and a minimum of approximately 10 fetal nucleated red blood cells (fnRBCs) starting from an initial 20 ml blood sample taken late in the first trimester.
  • WBCs maternal white blood cells
  • mnBCs maternal nuclear red blood cells
  • fnRBCs fetal nucleated red blood cells
  • STR loci multiple loci per chromosome of interest
  • STRs could be designed along chromosomes 13, 18, 21 and X to detect the most frequent aneuploidies, and along control regions of the genome where aneuploidy is not expected.
  • four or more STRs should be analyzed per chromosome of interest to ensure accurate detection of aneuploidy.
  • each primer can contain a common ⁇ 18b ⁇ sequence on the 5' end which can be used for the subsequent DNA cloning and sequencing procedures.
  • each well in the microtiter plate can be assigned a unique ⁇ 6bp DNA tag sequence which can be incorporated into the middle part of the upstream primer for each of the different
  • STRs The DNA tags make it possible to pool all of the STR amp Ikons following the multiplex PCR, which makes possible to analyze the amplicons in parallel during the ultra-deep sequencing procedure. Furthermore, nested PCR strategies for the STR amplification can achieve higher reliability of amplification from single cells. [00228] Following PCR, the amplicons from each of the wells in the microtiter plate are pooled, purified and analyzed using a single-molecule sequencing strategy such as the technology developed by 454 Life Sciences (Branford, CT). Briefly, the amplicons are diluted and mixed with beads such that each bead captures a single molecule of the amplified material.
  • Ultra-deep sequencing provides an accurate and quantitative way to measure the allele abundances for each of the STRs.
  • the total required number of reads for each of the aliquot wells is determined by the number of STRs and the error rates of the multiplex PCR and the Poisson sampling statistics associated with the sequencing procedures.
  • Statistical models which may account for variables in amplification can be used to detect ploidy changes with high levels of confidence. Using this statistical model it can be predicted that —100,000 to 300,000 sequence reads will be required to analyze each patient, with ⁇ 3 to 10 STR loci per chromosome.
  • Sequencing can be performed using the classic Sanger sequencing method or any other method known in the art.
  • sequencing can occur by sequencing-by-synthesis, which involves inferring the sequence of the template by synthesizing a strand complementary to the target nucleic acid sequence.
  • Sequence-by-synthesis can be initiated using sequencing primers complementary to the sequencing element on the nucleic acid tags.
  • the method involves detecting the identity of each nucleotide immediately after (substantially real-time) or upon (real-time) the incorporation of a labeled nucleotide or nucleotide analog into a growing strand of a complementary nucleic acid sequence in a polymerase reaction.
  • a signal is measured and then nulled by methods known in the art. Examples of sequence-by-synthesis methods are described in U.S. Application Publication Nos. 2003/0044781, 2006/0024711, 2006/0024678 and 2005/0100932.
  • labels that can be used to label nucleotide or nucleotide analogs for sequencing-by-synthesis include, but are not limited to, chromophores, fluorescent moieties, enzymes, antigens, heavy metal, magnetic probes, dyes, phosphorescent groups, radioactive materials, chemiluminescent moieties, scattering or fluorescent nanoparticles, Raman signal generating moieties, and electrochemical detection moieties.
  • Sequencing-by- synthesis can generate at least 1,000, at least 5,000, at least 10,000, at least 20,000, 30,000, at least 40,000, at least 50,000, at least 100,000 or at least 500,000 reads per hour.
  • Such reads can have at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 120 or at least 150 bases per read.
  • Another sequencing method involves hybridizing the amplified genomic region of interest to a primer complementary to it.
  • This hybridization complex is incubated with a polymerase, ATP sulfurylase, luciferase, apyrase, and the substrates luciferin and adenosine 5 ' phosphosulfate.
  • deoxynucleotide triphosphates corresponding to the bases A, C, G, and T (U) are added sequentially.
  • Each base incorporation is accompanied by release of pyrophosphate, converted to ATP by sulfurylase, which drives synthesis of oxyluciferin and the release of visible light.
  • Yet another sequencing method involves a four-color sequencing by ligation scheme (degenerate ligation), which involves hybridizing an anchor primer to one of four positions. Then an enzymatic ligation reaction of the anchor primer to a population of degenerate nonamers that are labeled with fluorescent dyes is performed. At any given cycle, the population of nonamers that is used is structure such that the identity of one of its positions is correlated with the identity of the fluorophore attached to that nonamer.
  • degenerate ligation involves hybridizing an anchor primer to one of four positions. Then an enzymatic ligation reaction of the anchor primer to a population of degenerate nonamers that are labeled with fluorescent dyes. At any given cycle, the population of nonamers that is used is structure such that the identity of one of its positions is correlated with the identity of the fluorophore attached to that nonamer.
  • the fluorescent signal allows the inference of the identity of the base.
  • the anchor prime ⁇ nonamer complexes are stripped and a new cycle begins.
  • Identify bins with non-maternal alleles e.g. fetal cells: The first step in the data analysis procedure would be to use the 6bp DNA tags to sort the 200,000 sequence reads into bins which correspond to the individual wells of the microtiter plates. The ⁇ 400 sequence reads from each of the bins would then be separated into the different STR groups using standard sequence alignment algorithms.
  • the aligned sequences from each of the bins would then be analyzed to identify non-maternal alleles.
  • These can be identified in one of two ways.
  • an independent blood sample fraction known to contain only maternal cells can be analyzed as described above. This sample can be a white blood cell fraction (which will contain only negligible numbers of fetal cells), or simply a dilution of the original sample before enrichment.
  • the genotype profiles for all the wells can be similarity-clustered to identify the dominant pattern associated with maternal cells. In either approach, the detection of non-maternal alleles then determines which wells in the initial microtiter plate contained fetal cells.
  • Determining the number bins with non-maternal alleles relative to the total number of bins provides an estimate of the number of fetal cells that were present in the original enriched cell population. Bins containing fetal cells would be identified with high levels of confidence because the non-maternal alleles are detected by multiple independent STRs.
  • Detect ploidy for chromosomes 13, 18, and 21 After identifying the bins that contained fetal cells, the next step would be to determine the ploidy of chromosomes 13, 18 and 21 by comparing the ratio of maternal to paternal alleles for each of the STRs. Again, for each bin there will be —33 sequence reads for each of the 12 STRs. In a normal fetus, a given STR will have 1:1 ratio of the maternal to paternal alleles with approximately 16 sequence reads corresponding to each allele (normal diallelic).
  • a trisomic fetus three doses of an STR marker can be detected either as three alleles with a 1:1:1 ratio (trisomic triallelic) or two alleles with a ratio of 2:1 (trisomic diallelic). In rare instances all three alleles may coincide and the locus will not be informative for that individual patient.
  • the information from the different STRs on each chromosome can be combined to increase the confidence of a given aneuploidy call.
  • the information from the independent bins containing fetal cells can also be combined to further increase the confidence of the call.
  • Microfluidic devices of the invention were designed by computer-aided design (CAD) and microfabricated by photolithography. A two-step process was developed in which a blood sample is first debulked to remove the large population of small cells, and then the rare target epithelial cells target cells are recovered by immuno affinity capture. The devices were defined by photolithography and etched into a silicon substrate based on the CAD-generated design.
  • the cell enrichment module which is approximately the size of a standard microscope slide, contains 14 parallel sample processing sections and associated sample handling channels that connect to common sample and buffer inlets and product and waste outlets. Each section contains an array of microfabricated obstacles that is optimized to enrich the target cell type by hydrodynamic size via displacement of the larger cells into the product stream.
  • the microchip was designed to separate red blood cells (RBCs) and platelets from the larger leukocytes and CTCs. Enriched populations of target cells were recovered from whole blood passed through the device. Performance of the cell enrichment microchip was evaluated by separating RBCs and platelets from white blood cells (WBCs) in normal whole blood (Figure 15). In cancer patients, CTCs are found in the larger
  • WBC fraction WBC fraction.
  • Blood was minimally diluted (30%), and a 6 ml sample was processed at a flow rate of up to 6 ml/hr.
  • the product and waste stream were evaluated in a Coulter Model "A -T diff ' clinical blood analyzer, which automatically distinguishes, sizes, and counts different blood cell populations.
  • the enrichment chip achieved separation of RBCs from WBCs, in which the WBC fraction had >99% retention of nucleated cells, >99% depletion of RBCs, and >97% depletion of platelets. Representative histograms of these cell fractions are shown in Figure 16. Routine cytology confirmed the high degree of enrichment of the WBC and RBC fractions (Figure 17).
  • epithelial cells were recovered by affinity capture in a microfluidic module that is functionalized with immobilized antibody.
  • a capture module with a single chamber containing a regular array of antibody-coated microfabricated obstacles was designed. These obstacles are disposed to maximize cell capture by increasing the capture area approximately four-fold, and by slowing the flow of cells under laminar flow adjacent to the obstacles to increase the contact time between the cells and the immobilized antibody.
  • the capture modules may be operated under conditions of relatively high flow rate but low shear to protect cells against damage.
  • the surface of the capture module was functionalized by sequential treatment with 10% silane, 0.5% gluteraldehyde, and avidin, followed by biotinylated anti- EpCAM.
  • CMRA reagent Cell Tracker Orange
  • Molecular Probes Eugene, OR
  • cell suspensions were processed directly in the capture modules without prior fractionation in the cell enrichment module to debulk the red blood cells; hence, the sample stream contained normal blood red cells and leukocytes as well as tumor cells.
  • the device was washed with buffer at a higher flow rate (3ml/hr) to remove the nonspecifically bound cells.
  • NCI-H 1650 cells that were spiked into whole blood and recovered by size fractionation and affinity capture as described above were successfully analyzed in situ.
  • a trial run to distinguish epithelial cells from leukocytes 0.5 ml of a stock solution of fluorescein-labeled CD45 pan-leukocyte monoclonal antibody were passed into the capture module and incubated at room temperature for 30 minutes, The module was washed with buffer to remove unbound antibody, and the cells were fixed on the chip with 1% paraformaldehyde and observed by fluorescence microscopy. As shown in Figure 18, the epithelial cells were bound to the obstacles and floor of the capture module. Background staining of the flow passages with CD45 pan-leukocyte antibody is visible, as are several stained leukocytes, apparently because of a low level of non-specific capture.
  • Example 6 Device embodiments
  • FIG. 19A A design for preferred device embodiments of the invention is shown in Figure 19A, and parameters corresponding to three preferred device embodiments associated with this design are shown in
  • FIGS. 19B and 19C These embodiments are particularly useful for enriching epithelial cells from blood.
  • Example 7 Determining counts for large cell types
  • a diagnosis of the absence, presence, or progression of cancer may be based on the number of cells in a cellular sample that are larger than a particular cutoff size. For example, cells with a hydrodynamic size of 14 ⁇ m or larger may be selected. This cutoff size would eliminate most leukocytes. The nature of these cells may then be determined by downstream molecular or cytological analysis.
  • Cell types other than epithelial cells that would be useful to analyze include endothelial cells, endothelial progenitor cells, endometrial cells, or trophoblasts indicative of a disease state. Furthermore, determining separate counts for epithelial cells, e.g., cancer cells, and other cell types, e.g., endothelial cells, followed by a determination of the ratios between the number of epithelial cells and the number of other cell types, may provide useful diagnostic information.
  • epithelial cells e.g., cancer cells
  • other cell types e.g., endothelial cells
  • a device of the invention may be configured to isolate targeted subpopulations of cells such as those described above, as shown in Figures 20A-D.
  • a size cutoff may be selected such that most native blood cells, including red blood cells, white blood cells, and platelets, flow to waste, while non-native cells, which could include endothelial cells, endothelial progenitor cells, endometrial cells, or trophoblasts, are collected in an enriched sample. This enriched sample may be further analyzed.
  • a device of the invention may include counting means to determine the number of cells in the enriched sample, or the number of cells of a particular type, e.g., cancer cells, within the enriched sample, and further analysis of the cells in the enriched sample may provide additional information that is useful for diagnostic or other purposes.
  • Example 8 Method for detection of EGFR mutations
  • a blood sample from a cancer patient is processed and analyzed using the devices and methods of the invention, resulting in an enriched sample of epithelial cells containing CTCs. This sample is then analyzed to identify potential EGFR mutations.
  • the method permits identification of both known, clinically relevant EGFR mutations, and discovery of novel mutations. An overview of this process is shown in Figure 22.
  • Sequence CTC EGFR mRNA a) Purify CTCs from blood sample; b) Purify total RNA from CTCs; c) Convert RNA to cDNA using reverse transcriptase; d) Use resultant cDNA to perform first and second PCR reactions for generating sequencing templates; and e) Purify the nested PCR amplicon and use as a sequencing template to sequence EGFR exons 18-21.
  • RNA sequencing 2) Confirm RNA sequence using CTC genomic DNA a) Purify CTCs from blood sample; b) Purify genomic DNA (gDNA) from CTCs; c) Amplify exons 18, 19, 20, and/or 21 via PCR reactions; and d) Use the resulting PCR amplicon(s) in real-time quantitative allele-specific PCR reactions in order to confirm the sequence of mutations discovered via RNA sequencing.
  • Sequence CTC EGFR mRNA a) Purify CTCs from blood sample. CTCs are isolated using any of the size-based enrichment and/or affinity purification devices of the invention. b) Purify total RNA from CTCs. Total RNA is then purified from isolated CTC populations using, e.g., the Qiagen Micro RNeasy kit, or a similar total RNA purification protocol from another manufacturer; alternatively, standard RNA purification protocols such as guanidium isothiocyanate homogenization followed by phenol/chloroform extraction and ethanol precipitation may be used. One such method is described in "Molecular Cloning — A Laboratory Manual, Second Edition" (1989) by J.
  • RNA to cDNA Convert RNA to cDNA using reverse transcriptase.
  • cDNA reactions are carried out based on the protocols of the supplier of reverse transcriptase. Typically, the amount of input RNA into the cDNA reactions is in the range of 10 picograms (pg) to 2 micrograms ( ⁇ g) total RNA.
  • First-strand DNA synthesis is carried out by hybridizing random 7mer DNA primers, or oligo-dT primers, or gene-specific primers, to RNA templates at 65°C followed by snap-chilling on ice.
  • cDNA synthesis is initiated by the addition of iScript Reverse Transcriptase (BioRad) or Superscript Reverse Transcriptase (Invitrogen) or a reverse transcriptase from another commercial vendor along with the appropriate enzyme reaction buffer.
  • iScript reverse transcriptase reactions are carried out at 42 0 C for 30-45 minutes, followed by enzyme inactivation for 5 minutes at 85°C.
  • cDNA is stored at -20 0 C until use or used immediately in PCR reactions.
  • cDNA reactions are carried out in a final volume of 20 ⁇ l, and 10% (2 ⁇ l) of the resultant cDNA is used in subsequent PCR reactions.
  • d) Use resultant cDNA to perform first and second PCR reactions for generating sequencing templates.
  • cDNA from the reverse transcriptase reactions is mixed with DNA primers specific for the region of interest ( Figure 23). See Table 3 for sets of primers that may be used for amplification of exons 18-21.
  • primer set M13(+)/M12(-) is internal to primer set Ml l(+)/M14(-).
  • primers M13(+) and M12(-) may be used in the nested round of amplification, if primers Ml 1(+) and M14(-) were used in the first round of expansion.
  • primer set Ml l(+)/M14(-) is internal to primer set M15(+)/M16(-)
  • primer set M23(+)/M24(-) is internal to primer set M21(+)/M22(-).
  • Hot Start PCR reactions are performed using Qiagen Hot- Star Taq Polymerase kit, or Applied Biosystems HotStart TaqMan polymerase, or other Hot Start thermostable polymerase, or without a hot start using Promega GoTaq Green Taq Polymerase master mix, TaqMan DNA polymerase, or other thermostable DNA polymerase.
  • reaction volumes are 50 ⁇ l
  • nucleotide triphosphates are present at a final concentration of 200 ⁇ M for each nucleotide
  • MgCl 2 is present at a final concentration of 1-4 mM
  • oligo primers are at a final concentration of 0.5 ⁇ M.
  • Hot start protocols begin with a 10-15 minute incubation at 95°C, followed by 40 cycles of 94°C for one minute (denaturation), 52°C for one minute (annealing), and 72°C for one minute (extension).
  • a lO minute terminal extension at 72°C is performed before samples are stored at 4°C until they are either used as template in the second (nested) round of PCRs, or purified using QiaQuick Spin Columns (Qiagen) prior to sequencing. If a hot-start protocol is not used, the initial incubation at 95°C is omitted. If a PCR product is to be used in a second round of PCRs, 2 ⁇ l (4%) of the initial PCR product is used as template in the second round reactions, and the identical reagent concentrations and cycling parameters are used.
  • PCR products are purified using Qiagen QuickSpin columns, the Agencourt AMPure PCR Purification System, or PCR product purification kits obtained from other vendors. After PCR products are purified, the nucleotide concentration and purity is determined with a Nanodrop 7000 spectrophotometer, and the PCR product concentration is brought to a concentration of 25 ng/ ⁇ l. As a quality control measure, only PCR products that have a UV-light absorbance ratio (A 26 (ZA 28O ) greater than 1.8 are used for sequencing. Sequencing primers are brought to a concentration of 3.2 pmol/ ⁇ l.
  • CTC genomic DNA is purified using the Qiagen DNeasy Mini kit, the Invirrogen ChargeSwitch gDNA kit, or another commercial kit, or via the following protocol:
  • Cell pellets are either lysed fresh or stored at -80 0 C and are thawed immediately before lysis.
  • Probe and primer sets are designed for all known mutations that affect gefitinib responsiveness in NSCLC patients, including over 40 such somatic mutations, including point mutations, deletions, and insertions, that have been reported in the medical literature.
  • somatic mutations including point mutations, deletions, and insertions
  • examples of primer and probe sets for five of the point mutations are listed in Table 5.
  • oligonucleotides may be designed using the primer optimization software program Primer Express (Applied Biosystems), with hybridization conditions optimized to distinguish the wild type EGFR DNA sequence from mutant alleles.
  • EGFR genomic DNA amplified from lung cancer cell lines that are known to carry EGFR mutations such as H358 (wild type), H1650 (15-bp deletion, ⁇ 2235-2249), and H1975 (two point mutations, 2369 C ⁇ T, 2573 T ⁇ G), is used to optimize the allele-specific Real Time PCR reactions.
  • H358 wild type
  • H1650 15-bp deletion, ⁇ 2235-2249
  • H1975 two point mutations, 2369 C ⁇ T, 2573 T ⁇ G
  • labeled probes containing wild type sequence are multiplexed in the same reaction with a single mutant probe. Expressing the results as a ratio of one mutant allele sequence versus wild type sequence may identify samples containing or lacking a given mutation. After conditions are optimized for a given probe set, it is then possible to multiplex probes for all of the mutant alleles withm a given exon within the same Real Time PCR assay, increasing the ease of use of this analytical tool in clinical settings.
  • a unique probe is designed for each wild type allele and mutant allele sequence. Wild-type sequences are marked with the fluorescent dye VIC at the 5' end, and mutant sequences with the fluorophore FAM.
  • a fluorescence quencher and Minor Groove Binding moiety are attached to the 3' ends of the probes.
  • ROX is used as a passive reference dye for normalization purposes.
  • a standard curve is generated for wild type sequences and is used for relative quantitation. Precise quantitation of mutant signal is not required, as the input cell population is of unknown, and varying, purity.
  • the assay is set up as described by ABI product literature, and the presence of a mutation is confirmed when the signal from a mutant allele probe rises above the background level of fluorescence (Figure 25), and this threshold cycle gives the relative frequency of the mutant allele in the input sample.
  • BCKDK branched-chain a-ketoacid dehydrogenase complex kinase
  • CD45 - specifically expressed in leukocytes a positive control for leukocytes and a negative control for tumor cells
  • EpCaM - specifically expressed in epithelial cells a negative control for leukocytes and a positive control for tumor cells
  • RNAs of approximately 9x10 6 leukocytes isolated using a cell enrichment device of the invention (cutoff size 4 ⁇ m) and 5 ⁇ lO s H1650 cells were isolated by using RNeasy mini kit (Qiagen). Two micrograms of total RNAs from leukocytes and Hl 650 cells were reverse transcribed to obtain first strand cDNAs using 100 pmol random hexamer (Roche) and 200 U Superscript II (Invitrogen) in a 20 ⁇ l reaction. The subsequent PCR was carried out using 0.5 ⁇ l of the first strand cDNA reaction and 10 pmol of forward and reverse primers in total 25 ⁇ l of mixture.
  • PCR was run for 40 cycles of 95°C for 20 seconds, 56°C for 20 seconds, and 70 0 C for 30 seconds.
  • the amplified products were separated on a 1% agarose gel.
  • BCKDK was found to be expressed in both leukocytes and H1650 cells; CD45 was expressed only in leukocytes; and both EpCAM and EGFR were expressed only in H 1650 cells.
  • Example 6 EGFR assay with low quantities of target RNA or high quantities of background RNA
  • various quantities of input NSCLC cell line total RNA were tested, ranging from 100 pg to 50 ng.
  • the results of the first and second EGFR PCR reactions are shown in Figure 27.
  • the first PCR reaction was shown to be sufficiently sensitive to detect 1 ng of input RNA, while the second round increased the sensitivity to 100 pg or less of input RNA. This corresponds to 7-10 cells, demonstrating that even extremely dilute samples may generate detectable signals using this assay.
  • Table 7 lists the RNA yield in a variety of cells and shows that the yield per cell is widely variable, depending on the cell type. This information is useful in order to estimate the amount of target and background RNA in a sample based on cell counts. For example, 1 ng of NCL-H 1975 RNA corresponds to approximately 100 cells, while 1 ⁇ g of PBMC RNA corresponds to approximately 10 6 cells. Thus, the highest contamination level in the above-described experiment, 1,000:1 of PBMC RNA to NCL- H1975 RNA, actually corresponds to a 10,000:1 ratio of PBMCs to NCL-H1975 cells. Thus, these data indicate that EGFR may be sequenced from as few as 100 CTCs contaminated by as many as 10 5 leukocytes.
  • RNAs were DNase I-treated for 30 minutes and then extracted with phenol/chloroform and precipitated with ethanol prior to first strand cDNA synthesis and subsequent PCR amplification. These steps were repeated with a second blood sample and a second chip.
  • the cDNA synthesized from chipl and chip2 RNAs along with H 1650 and leukocyte cDNAs were PCR amplified using two sets of primers, CD45 1 (SEQ ID NO:45) and CD45 2 (SEQ ID NO:46) (Table 6) as well as EGFR_5 (forward primer, S'-GTTCGGCACGGTGTATAAGG-S') (SEQ ID NO: 52) and EGFR_6 (reverse primer, 5'-CTGGCCATCACGTAGGCTTC-S') (SEQ ID NO:53).
  • EGFR 5 and EGFR_6 produce a 138 bp wild type amplified fragment and a 123 bp mutant amplified fragment in H1650 cells.
  • PCR products were separated on a 2.5% agarose gel. As shown in Figure 29, EGFR wild type and mutant amplified fragments were readily detected, despite the high leukocyte background, demonstrating that the EGFR assay is robust and does not require a highly purified sample.

Abstract

L'invention concerne des méthodes de diagnostic ou de pronostic d'un cancer chez un sujet par enrichissement, détection et analyse de cellules rares individuelles, p. ex. de cellules épithéliales, dans un échantillon prélevé sur un sujet. L'invention concerne également des procédés de marquage de régions de l'ADN génomique dans les cellules individuelles présentes dans ledit échantillon mixte, au moyen de différents marqueurs, chaque marqueur étant spécifique de chaque cellule, et de quantification des régions d'ADN génomique marquées de chaque cellule de l'échantillon mixte. Ce procédé consiste plus particulièrement à détecter la présence de mutations géniques dans les cellules individuelles d'un sous-échantillon.
PCT/US2007/071148 2006-06-14 2007-06-13 Analyse de cellules rares par division d'échantillon et utilisation de marqueurs d'adn WO2008111990A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US80481706P 2006-06-14 2006-06-14
US80481906P 2006-06-14 2006-06-14
US60/804,817 2006-06-14
US60/804,819 2006-06-14

Publications (1)

Publication Number Publication Date
WO2008111990A1 true WO2008111990A1 (fr) 2008-09-18

Family

ID=38832121

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2007/071148 WO2008111990A1 (fr) 2006-06-14 2007-06-13 Analyse de cellules rares par division d'échantillon et utilisation de marqueurs d'adn
PCT/US2007/071149 WO2007147018A1 (fr) 2006-06-14 2007-06-13 Analyse d'échantillons enrichis de cellules rares

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2007/071149 WO2007147018A1 (fr) 2006-06-14 2007-06-13 Analyse d'échantillons enrichis de cellules rares

Country Status (2)

Country Link
US (4) US20080124721A1 (fr)
WO (2) WO2008111990A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9493831B2 (en) 2010-01-23 2016-11-15 Verinata Health, Inc. Methods of fetal abnormality detection
CN107208164A (zh) * 2015-02-13 2017-09-26 泊克玛科技有限公司 用于对循环肿瘤细胞进行测序的血液样品处理
US10324011B2 (en) 2013-03-15 2019-06-18 The Trustees Of Princeton University Methods and devices for high throughput purification
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
US11142746B2 (en) 2013-03-15 2021-10-12 University Of Maryland, Baltimore High efficiency microfluidic purification of stem cells to improve transplants
US11268091B2 (en) 2018-12-13 2022-03-08 Dna Script Sas Direct oligonucleotide synthesis on cells and biomolecules
US11493428B2 (en) 2013-03-15 2022-11-08 Gpb Scientific, Inc. On-chip microfluidic processing of particles

Families Citing this family (141)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2359689B1 (fr) 2002-09-27 2015-08-26 The General Hospital Corporation Dispositif microfluidique pour la séparation de cellules et usage du dispositif
ITBO20040420A1 (it) 2004-07-07 2004-10-07 Type S R L Macchina per taglio e formatura di piattine metalliche
US20070026418A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
EP1915618A4 (fr) * 2005-06-02 2009-09-30 Fluidigm Corp Analyse utilisant des dispositifs de separation microfluidiques
ITBO20050481A1 (it) 2005-07-19 2007-01-20 Silicon Biosystems S R L Metodo ed apparato per la manipolazione e/o l'individuazione di particelle
US11111544B2 (en) 2005-07-29 2021-09-07 Natera, Inc. System and method for cleaning noisy genetic data and determining chromosome copy number
US9424392B2 (en) 2005-11-26 2016-08-23 Natera, Inc. System and method for cleaning noisy genetic data from target individuals using genetic data from genetically related individuals
US8921102B2 (en) 2005-07-29 2014-12-30 Gpb Scientific, Llc Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US11111543B2 (en) 2005-07-29 2021-09-07 Natera, Inc. System and method for cleaning noisy genetic data and determining chromosome copy number
US10081839B2 (en) 2005-07-29 2018-09-25 Natera, Inc System and method for cleaning noisy genetic data and determining chromosome copy number
ITBO20050646A1 (it) 2005-10-26 2007-04-27 Silicon Biosystem S R L Metodo ed apparato per la caratterizzazione ed il conteggio di particelle
EP3591068A1 (fr) 2006-02-02 2020-01-08 The Board of Trustees of the Leland Stanford Junior University Dépistage génétique non invasif du f tus par analyse numérique
ITTO20060226A1 (it) 2006-03-27 2007-09-28 Silicon Biosystem S P A Metodo ed apparato per il processamento e o l'analisi e o la selezione di particelle, in particolare particelle biologiche
US8137912B2 (en) * 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
EP2589668A1 (fr) 2006-06-14 2013-05-08 Verinata Health, Inc Analyse de cellules rares utilisant la division d'échantillons et les marqueurs d'ADN
US20080124721A1 (en) * 2006-06-14 2008-05-29 Martin Fuchs Analysis of rare cell-enriched samples
EP2527471B1 (fr) 2007-07-23 2020-03-04 The Chinese University of Hong Kong Diagnostic du cancer utilisant un séquençage génomique
US20100112590A1 (en) 2007-07-23 2010-05-06 The Chinese University Of Hong Kong Diagnosing Fetal Chromosomal Aneuploidy Using Genomic Sequencing With Enrichment
ITTO20070771A1 (it) 2007-10-29 2009-04-30 Silicon Biosystems Spa Metodo e apparato per la identificazione e manipolazione di particelle
US8008032B2 (en) * 2008-02-25 2011-08-30 Cellective Dx Corporation Tagged ligands for enrichment of rare analytes from a mixed sample
US9328172B2 (en) * 2008-04-05 2016-05-03 Single Cell Technology, Inc. Method of obtaining antibodies of interest and nucleotides encoding same
US10429376B2 (en) 2008-05-16 2019-10-01 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Microfluidic isolation of tumor cells or other rare cells from whole blood or other liquids
EP2318552B1 (fr) 2008-09-05 2016-11-23 TOMA Biosciences, Inc. Procédés pour la stratification et l'annotation des options de traitement médicamenteux contre le cancer
HUE031849T2 (en) 2008-09-20 2017-08-28 Univ Leland Stanford Junior Non-invasive diagnosis of fetal aneuploidy by sequencing
JP2012504956A (ja) * 2008-10-10 2012-03-01 セントレ ナショナル デ ラ レシェルシェ サイエンティフィーク−ディーエーイー 細胞ソート・デバイス
IT1391619B1 (it) * 2008-11-04 2012-01-11 Silicon Biosystems Spa Metodo per l'individuazione, selezione e analisi di cellule tumorali
US10895575B2 (en) 2008-11-04 2021-01-19 Menarini Silicon Biosystems S.P.A. Method for identification, selection and analysis of tumour cells
EP2376659B1 (fr) * 2008-12-17 2015-12-02 Life Technologies Corporation Procédés, compositions et nécessaires de détection de variantes alléliques
CA2748030A1 (fr) 2008-12-22 2010-07-01 Arnold R. Oliphant Procedes et listes de genotypage permettant de detecter des alleles, des genomes et des transcriptomes
US8162149B1 (en) 2009-01-21 2012-04-24 Sandia Corporation Particle sorter comprising a fluid displacer in a closed-loop fluid circuit
WO2010085815A1 (fr) * 2009-01-26 2010-07-29 Artemis Health, Inc. Méthodes et compositions d'identification d'une cellule foetale
SG174459A1 (en) 2009-03-17 2011-10-28 Silicon Biosystems Spa Microfluidic device for isolation of cells
JP5805064B2 (ja) 2009-03-27 2015-11-04 ライフ テクノロジーズ コーポレーション 対立遺伝子変種を検出するための方法、組成物、およびキット
AU2010232439C1 (en) 2009-04-02 2017-07-13 Fluidigm Corporation Multi-primer amplification method for barcoding of target nucleic acids
WO2010124227A2 (fr) * 2009-04-24 2010-10-28 The Board Of Trustees Of The University Of Illinois Procédés et dispositifs de capture de cellules tumorales circulantes
CA2760439A1 (fr) 2009-04-30 2010-11-04 Good Start Genetics, Inc. Procedes et compositions d'evaluation de marqueurs genetiques
US8825412B2 (en) 2010-05-18 2014-09-02 Natera, Inc. Methods for non-invasive prenatal ploidy calling
EP2309273B1 (fr) * 2009-09-16 2016-05-18 ZEILLINGER, Robert Détermination de nouveau marqueur de tumeur
EP2473638B1 (fr) 2009-09-30 2017-08-09 Natera, Inc. Méthode non invasive de détermination d'une ploïdie prénatale
AU2010310688A1 (en) 2009-10-21 2012-05-17 Epic Sciences, Inc. Method of using non-rare cells to detect rare cells
CN102762712A (zh) 2010-01-21 2012-10-31 百赛普有限公司 稀有细胞的磁性分离
US11332793B2 (en) 2010-05-18 2022-05-17 Natera, Inc. Methods for simultaneous amplification of target loci
US11339429B2 (en) 2010-05-18 2022-05-24 Natera, Inc. Methods for non-invasive prenatal ploidy calling
US11326208B2 (en) 2010-05-18 2022-05-10 Natera, Inc. Methods for nested PCR amplification of cell-free DNA
US11322224B2 (en) 2010-05-18 2022-05-03 Natera, Inc. Methods for non-invasive prenatal ploidy calling
US11332785B2 (en) 2010-05-18 2022-05-17 Natera, Inc. Methods for non-invasive prenatal ploidy calling
US11939634B2 (en) 2010-05-18 2024-03-26 Natera, Inc. Methods for simultaneous amplification of target loci
US11408031B2 (en) 2010-05-18 2022-08-09 Natera, Inc. Methods for non-invasive prenatal paternity testing
US20190010543A1 (en) 2010-05-18 2019-01-10 Natera, Inc. Methods for simultaneous amplification of target loci
US10316362B2 (en) 2010-05-18 2019-06-11 Natera, Inc. Methods for simultaneous amplification of target loci
US9677118B2 (en) 2014-04-21 2017-06-13 Natera, Inc. Methods for simultaneous amplification of target loci
US20120034603A1 (en) * 2010-08-06 2012-02-09 Tandem Diagnostics, Inc. Ligation-based detection of genetic variants
US20140342940A1 (en) 2011-01-25 2014-11-20 Ariosa Diagnostics, Inc. Detection of Target Nucleic Acids using Hybridization
US8700338B2 (en) 2011-01-25 2014-04-15 Ariosa Diagnosis, Inc. Risk calculation for evaluation of fetal aneuploidy
US11203786B2 (en) 2010-08-06 2021-12-21 Ariosa Diagnostics, Inc. Detection of target nucleic acids using hybridization
US20130261003A1 (en) * 2010-08-06 2013-10-03 Ariosa Diagnostics, In. Ligation-based detection of genetic variants
US10533223B2 (en) 2010-08-06 2020-01-14 Ariosa Diagnostics, Inc. Detection of target nucleic acids using hybridization
US10167508B2 (en) * 2010-08-06 2019-01-01 Ariosa Diagnostics, Inc. Detection of genetic abnormalities
US20130040375A1 (en) * 2011-08-08 2013-02-14 Tandem Diagnotics, Inc. Assay systems for genetic analysis
AU2015201176B2 (en) * 2010-08-06 2017-09-07 F. Hoffmann-La Roche Ag Assay systems for determination of source contribution in a sample
CA2810931C (fr) 2010-09-24 2018-04-17 The Board Of Trustees Of The Leland Stanford Junior University Capture directe, amplification et sequencage d'adn cible a l'aide d'amorces immobilisees
CA2814049C (fr) * 2010-10-08 2021-07-13 President And Fellows Of Harvard College Etablissement a haut debit d'un code-barres de cellules simples
WO2012068243A2 (fr) * 2010-11-16 2012-05-24 Ingeneron, Inc. Procédés pour obtenir des cellules cibles
EP2656263B1 (fr) 2010-12-22 2019-11-06 Natera, Inc. Procédés de recherche de paternité prénatale, non invasive
IT1403518B1 (it) 2010-12-22 2013-10-31 Silicon Biosystems Spa Dispositivo microfluidico per la manipolazione di particelle
US9163281B2 (en) 2010-12-23 2015-10-20 Good Start Genetics, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
US10131947B2 (en) * 2011-01-25 2018-11-20 Ariosa Diagnostics, Inc. Noninvasive detection of fetal aneuploidy in egg donor pregnancies
JP6153874B2 (ja) 2011-02-09 2017-06-28 ナテラ, インコーポレイテッド 非侵襲的出生前倍数性呼び出しのための方法
US20120238458A1 (en) * 2011-03-16 2012-09-20 Dr. Robert Zeillinger Novel tumor marker determination
CN110441343A (zh) 2011-03-22 2019-11-12 通用医疗公司 利用磁共振检测目的物
WO2012129363A2 (fr) 2011-03-24 2012-09-27 President And Fellows Of Harvard College Détection et analyse d'acide nucléique d'une cellule isolée
WO2012162267A2 (fr) * 2011-05-20 2012-11-29 Fluidigm Corporation Réactions d'encodage d'acide nucléique
US9926552B2 (en) * 2011-06-06 2018-03-27 Cornell University Microfluidic device for extracting, isolating, and analyzing DNA from cells
US9541480B2 (en) 2011-06-29 2017-01-10 Academia Sinica Capture, purification, and release of biological substances using a surface coating
JP5659319B2 (ja) 2011-06-29 2015-01-28 ビージーアイ ヘルス サービス カンパニー リミテッド 胎児の遺伝的異常の非侵襲的検出
US10466160B2 (en) * 2011-08-01 2019-11-05 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
WO2013019491A1 (fr) 2011-08-01 2013-02-07 Denovo Sciences Système de capture de cellules et procédé d'utilisation
US9404864B2 (en) 2013-03-13 2016-08-02 Denovo Sciences, Inc. System for imaging captured cells
US9228233B2 (en) 2011-10-17 2016-01-05 Good Start Genetics, Inc. Analysis methods
ITTO20110990A1 (it) 2011-10-28 2013-04-29 Silicon Biosystems Spa Metodo ed apparato per l'analisi ottica di particelle a basse temperature
ITBO20110766A1 (it) 2011-12-28 2013-06-29 Silicon Biosystems Spa Dispositivi, apparato, kit e metodo per il trattamento di un campione biologico
US8209130B1 (en) 2012-04-04 2012-06-26 Good Start Genetics, Inc. Sequence assembly
US10227635B2 (en) 2012-04-16 2019-03-12 Molecular Loop Biosolutions, Llc Capture reactions
SG11201407901PA (en) 2012-05-21 2015-01-29 Fluidigm Corp Single-particle analysis of particle populations
US11913065B2 (en) 2012-09-04 2024-02-27 Guardent Health, Inc. Systems and methods to detect rare mutations and copy number variation
US20160040229A1 (en) 2013-08-16 2016-02-11 Guardant Health, Inc. Systems and methods to detect rare mutations and copy number variation
US10876152B2 (en) 2012-09-04 2020-12-29 Guardant Health, Inc. Systems and methods to detect rare mutations and copy number variation
KR102393608B1 (ko) 2012-09-04 2022-05-03 가던트 헬쓰, 인크. 희귀 돌연변이 및 카피수 변이를 검출하기 위한 시스템 및 방법
US9494500B2 (en) * 2012-10-29 2016-11-15 Academia Sinica Collection and concentration system for biologic substance of interest and use thereof
US9752181B2 (en) 2013-01-26 2017-09-05 Denovo Sciences, Inc. System and method for capturing and analyzing cells
EP2971159B1 (fr) 2013-03-14 2019-05-08 Molecular Loop Biosolutions, LLC Procédés d'analyse d'acides nucléiques
GB201304810D0 (en) * 2013-03-15 2013-05-01 Isis Innovation Assay
US10391490B2 (en) 2013-05-31 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US9856535B2 (en) 2013-05-31 2018-01-02 Denovo Sciences, Inc. System for isolating cells
US10577655B2 (en) 2013-09-27 2020-03-03 Natera, Inc. Cell free DNA diagnostic testing standards
US10262755B2 (en) 2014-04-21 2019-04-16 Natera, Inc. Detecting cancer mutations and aneuploidy in chromosomal segments
US10851414B2 (en) 2013-10-18 2020-12-01 Good Start Genetics, Inc. Methods for determining carrier status
WO2015095603A1 (fr) 2013-12-19 2015-06-25 Axon Dx, Llc Procédés et appareil de détection, capture et isolement de cellules
CN106062214B (zh) 2013-12-28 2020-06-09 夸登特健康公司 用于检测遗传变异的方法和系统
WO2015112914A1 (fr) 2014-01-24 2015-07-30 Life Technologies Corporation Épuration de produits chimiques et formats pour des réactions de séquençage d'adn de sanger sur un dispositif micro-fluidique
EP3100047B1 (fr) 2014-01-27 2021-08-11 Epic Sciences, Inc. Diagnostics de cellules tumorales circulantes pour biomarqueurs du cancer de la prostate
RS61976B1 (sr) * 2014-02-18 2021-07-30 Illumina Inc Postupci i kompozicije za određivanje dnk profila
WO2015127008A1 (fr) 2014-02-21 2015-08-27 Epic Sciences, Inc. Méthodes pour analyser des cellules circulantes rares
WO2015134972A2 (fr) 2014-03-07 2015-09-11 The Board Of Trustees Of The University Of Illinois Dispositif microfluidique biomimétique destiné à la capture de cellules tumorales circulantes
EP3126814B1 (fr) 2014-04-01 2019-06-12 Academia Sinica Procédés et systèmes pour le diagnostic et le pronostic du cancer
RU2717641C2 (ru) 2014-04-21 2020-03-24 Натера, Инк. Обнаружение мутаций и плоидности в хромосомных сегментах
US11053548B2 (en) 2014-05-12 2021-07-06 Good Start Genetics, Inc. Methods for detecting aneuploidy
ES2872344T3 (es) * 2014-07-02 2021-11-02 Siemens Healthcare Diagnostics Inc Separación selectiva de ácidos nucleicos
US10112198B2 (en) 2014-08-26 2018-10-30 Academia Sinica Collector architecture layout design
WO2016040446A1 (fr) 2014-09-10 2016-03-17 Good Start Genetics, Inc. Procédés permettant la suppression sélective de séquences non cibles
EP3224595A4 (fr) 2014-09-24 2018-06-13 Good Start Genetics, Inc. Commande de procédé pour accroître la robustesse de tests génétiques
WO2016112073A1 (fr) 2015-01-06 2016-07-14 Good Start Genetics, Inc. Criblage de variants structuraux
US10364467B2 (en) 2015-01-13 2019-07-30 The Chinese University Of Hong Kong Using size and number aberrations in plasma DNA for detecting cancer
US11479812B2 (en) 2015-05-11 2022-10-25 Natera, Inc. Methods and compositions for determining ploidy
US10976232B2 (en) 2015-08-24 2021-04-13 Gpb Scientific, Inc. Methods and devices for multi-step cell purification and concentration
CA3006994A1 (fr) 2015-12-16 2017-06-22 Fluidigm Corporation Amplification multiplex de haut niveau
WO2017106768A1 (fr) 2015-12-17 2017-06-22 Guardant Health, Inc. Procédés de détermination du nombre de copies du gène tumoral par analyse d'adn acellulaire
US10107726B2 (en) 2016-03-16 2018-10-23 Cellmax, Ltd. Collection of suspended cells using a transferable membrane
US11383240B2 (en) 2016-05-22 2022-07-12 Cornell University Single cell whole genome amplification via micropillar arrays under flow conditions
EP3518974A4 (fr) 2016-09-29 2020-05-27 Myriad Women's Health, Inc. Dépistage prénatal non invasif utilisant une optimisation de profondeur itérative dynamique
US9850523B1 (en) 2016-09-30 2017-12-26 Guardant Health, Inc. Methods for multi-resolution analysis of cell-free nucleic acids
KR20210158870A (ko) 2016-09-30 2021-12-31 가던트 헬쓰, 인크. 무세포 핵산의 다중-해상도 분석 방법
US11485996B2 (en) 2016-10-04 2022-11-01 Natera, Inc. Methods for characterizing copy number variation using proximity-litigation sequencing
US10011870B2 (en) 2016-12-07 2018-07-03 Natera, Inc. Compositions and methods for identifying nucleic acid molecules
CA3049139A1 (fr) 2017-02-21 2018-08-30 Natera, Inc. Compositions, procedes, et kits d'isolement d'acides nucleiques
US11453005B2 (en) 2017-05-25 2022-09-27 Rutgers, The State University Of New Jersey Anchored-liquid stationary phase for separation and filtration systems
EP3651903A4 (fr) 2017-08-29 2021-06-16 Bio-Rad Laboratories, Inc. Système et procédé pour isoler et analyser des cellules
AU2018323875A1 (en) 2017-09-01 2020-04-23 Gpb Scientific, Inc. Methods for preparing therapeutically active cells using microfluidics
US11525159B2 (en) 2018-07-03 2022-12-13 Natera, Inc. Methods for detection of donor-derived cell-free DNA
JP2022519045A (ja) 2019-01-31 2022-03-18 ガーダント ヘルス, インコーポレイテッド 無細胞dnaを単離するための組成物および方法
DE102019202788A1 (de) * 2019-03-01 2020-09-03 Robert Bosch Gmbh Verfahren zum Zählen von Zelltypen oder Zellmarkern in einer Probe, insbesondere in einer Blutprobe
US10633693B1 (en) 2019-04-16 2020-04-28 Celsee Diagnostics, Inc. System and method for leakage control in a particle capture system
US11273439B2 (en) 2019-05-07 2022-03-15 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
KR20220016477A (ko) 2019-05-07 2022-02-09 바이오 래드 래버러토리스 인코오포레이티드 자동화된 단일 세포 처리를 위한 시스템 및 방법
SG11202112898WA (en) 2019-06-14 2021-12-30 Bio Rad Laboratories System and method for automated single cell processing and analyses
EP3999081A1 (fr) 2019-07-18 2022-05-25 GPB Scientific, Inc. Traitement ordonné de produits sanguins pour produire des cellules thérapeutiquement actives
CA3166192A1 (fr) 2019-12-28 2021-07-01 Gpb Scientific, Inc. Cartouches microfluidiques pour le traitement de particules et de cellules
WO2021154965A1 (fr) * 2020-01-31 2021-08-05 Luna Genetics, Inc. Détermination de génotype foetal à l'aide d'un échantillon biologique maternel
US11504719B2 (en) 2020-03-12 2022-11-22 Bio-Rad Laboratories, Inc. System and method for receiving and delivering a fluid for sample processing
GB2598272A (en) * 2020-03-19 2022-03-02 Schreier Stefan Means and method for detecting aberrant erythroblasts
CN114628370A (zh) * 2020-12-11 2022-06-14 联华电子股份有限公司 半导体结构

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004113877A1 (fr) * 2003-06-13 2004-12-29 The General Hospital Corporation Systemes microfluidiques d'elimination basee sur la taille de globules rouges et de plaquettes du sang
US20060073125A1 (en) * 2000-08-03 2006-04-06 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US20060223178A1 (en) * 2005-04-05 2006-10-05 Tom Barber Devices and methods for magnetic enrichment of cells and other particles

Family Cites Families (117)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3560754A (en) * 1965-11-17 1971-02-02 Ibm Photoelectric particle separator using time delay
US5310674A (en) * 1982-05-10 1994-05-10 Bar-Ilan University Apertured cell carrier
US4508625A (en) * 1982-10-18 1985-04-02 Graham Marshall D Magnetic separation using chelated magnetic ions
US5164598A (en) * 1985-08-05 1992-11-17 Biotrack Capillary flow device
US5721098A (en) * 1986-01-16 1998-02-24 The Regents Of The University Of California Comparative genomic hybridization
US4936465A (en) * 1987-12-07 1990-06-26 Zoeld Tibor Method and apparatus for fast, reliable, and environmentally safe dispensing of fluids, gases and individual particles of a suspension through pressure control at well defined parts of a closed flow-through system
US5215926A (en) * 1988-06-03 1993-06-01 Cellpro, Inc. Procedure for designing efficient affinity cell separation processes
US5641628A (en) * 1989-11-13 1997-06-24 Children's Medical Center Corporation Non-invasive method for isolation and detection of fetal DNA
US5622831A (en) * 1990-09-26 1997-04-22 Immunivest Corporation Methods and devices for manipulation of magnetically collected material
US5296375A (en) * 1992-05-01 1994-03-22 Trustees Of The University Of Pennsylvania Mesoscale sperm handling devices
US5304487A (en) * 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5486335A (en) * 1992-05-01 1996-01-23 Trustees Of The University Of Pennsylvania Analysis based on flow restriction
US5498392A (en) * 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
US5637469A (en) * 1992-05-01 1997-06-10 Trustees Of The University Of Pennsylvania Methods and apparatus for the detection of an analyte utilizing mesoscale flow systems
US5726026A (en) * 1992-05-01 1998-03-10 Trustees Of The University Of Pennsylvania Mesoscale sample preparation device and systems for determination and processing of analytes
US5629147A (en) * 1992-07-17 1997-05-13 Aprogenex, Inc. Enriching and identifying fetal cells in maternal blood for in situ hybridization
DE4244715C2 (de) * 1992-08-26 1995-06-14 Kuebler Gmbh Dr Verfahren zur Herstellung individuum-spezifischer, gegen Tumorantigene gerichtete monoklonale Antikörper
WO1994007138A1 (fr) * 1992-09-14 1994-03-31 Fodstad Oystein Detection de cellules cibles specifiques dans une population de cellules specialisees ou mixtes et solutions contenant des populations de cellules mixtes
US5489506A (en) * 1992-10-26 1996-02-06 Biolife Systems, Inc. Dielectrophoretic cell stream sorter
US5427663A (en) * 1993-06-08 1995-06-27 British Technology Group Usa Inc. Microlithographic array for macromolecule and cell fractionation
US5556773A (en) * 1993-08-06 1996-09-17 Yourno; Joseph Method and apparatus for nested polymerase chain reaction (PCR) with single closed reaction tubes
US5840502A (en) * 1994-08-31 1998-11-24 Activated Cell Therapy, Inc. Methods for enriching specific cell-types by density gradient centrifugation
US5662813A (en) * 1994-10-21 1997-09-02 Bioseparations, Inc. Method for separation of nucleated fetal erythrocytes from maternal blood samples
US5750339A (en) * 1994-11-30 1998-05-12 Thomas Jefferson University Methods for identifying fetal cells
US6190870B1 (en) * 1995-08-28 2001-02-20 Amcell Corporation Efficient enrichment and detection of disseminated tumor cells
US6718053B1 (en) * 1996-11-27 2004-04-06 Chromavision Medical Systems, Inc. Method and apparatus for automated image analysis of biological specimens
US6215892B1 (en) * 1995-11-30 2001-04-10 Chromavision Medical Systems, Inc. Method and apparatus for automated image analysis of biological specimens
US5863502A (en) * 1996-01-24 1999-01-26 Sarnoff Corporation Parallel reaction cassette and associated devices
US5891651A (en) * 1996-03-29 1999-04-06 Mayo Foundation For Medical Education And Research Methods of recovering colorectal epithelial cells or fragments thereof from stool
US6387707B1 (en) * 1996-04-25 2002-05-14 Bioarray Solutions Array Cytometry
US5989835A (en) * 1997-02-27 1999-11-23 Cellomics, Inc. System for cell-based screening
JP3996644B2 (ja) * 1996-06-07 2007-10-24 イムニベスト・コーポレイション 外部及び内部勾配を有する磁気分離
US6074827A (en) * 1996-07-30 2000-06-13 Aclara Biosciences, Inc. Microfluidic method for nucleic acid purification and processing
WO1998010267A1 (fr) * 1996-09-04 1998-03-12 Technical University Of Denmark Systeme a microdebit pour separation et analyse de particules
US5879624A (en) * 1997-01-15 1999-03-09 Boehringer Laboratories, Inc. Method and apparatus for collecting and processing blood
SK118099A3 (en) * 1997-02-28 2000-05-16 Burstein Lab Inc Laboratory in a disk
US7214298B2 (en) * 1997-09-23 2007-05-08 California Institute Of Technology Microfabricated cell sorter
US6540895B1 (en) * 1997-09-23 2003-04-01 California Institute Of Technology Microfabricated cell sorter for chemical and biological materials
US6111096A (en) * 1997-10-31 2000-08-29 Bbi Bioseq, Inc. Nucleic acid isolation and purification
CA2318880C (fr) * 1998-02-04 2010-07-27 Perkin-Elmer Corporation Determination d'un genotype d'un produit d'amplification sur des sites alleliques multiples
BR9907852A (pt) * 1998-02-12 2000-10-24 Immunivest Corp Processos para detectar e enumerar células raras e cancerosas em uma população celular mista, para diagnosticar câncer de estágio precoce em um paciente de teste, para determinar a probabilidade de recorrência de câncer em um paciente humano anteriormente tratado de câncer, para distinguir um carcinoma confinado ao órgão de um carcinoma com propriedades metastáticas, para acompanhar a situação de remissão em um paciente humano com câncer que passa pelo tratamento de terapia contra o câncer e para aumentar quantidades de células epiteliais circulantes em uma amostra de sangue, partìcula magnética revestida, composição, conjuntos de teste para avaliar uma amostra de paciente quanto a presença de células raras circulantes, quanto a presença de células de tumor circulantes, quanto a presença de células de câncer de mama circulantes, quanto a presença de células de câncer de próstata circulantes, quanto a presença de células de câncer de cólon circulantes, quanto a presença de células de câncer de bexiga circulantes e para monitorar um paciente quanto a recorrência de câncer, e, fração de sangue periférico enriquecido quanto a células neoplásticas circulantes
US6036857A (en) * 1998-02-20 2000-03-14 Florida State University Research Foundation, Inc. Apparatus for continuous magnetic separation of components from a mixture
US6210910B1 (en) * 1998-03-02 2001-04-03 Trustees Of Tufts College Optical fiber biosensor array comprising cell populations confined to microcavities
US6200765B1 (en) * 1998-05-04 2001-03-13 Pacific Northwest Cancer Foundation Non-invasive methods to detect prostate cancer
US6606413B1 (en) * 1998-06-01 2003-08-12 Trestle Acquisition Corp. Compression packaged image transmission for telemicroscopy
FR2782730B1 (fr) * 1998-08-25 2002-05-17 Biocom Sa Procede de separation cellulaire pour l'isolation de cellules pathogeniques, notamment cancereuses rares, equipement et reactif pour la mise en oeuvre du procede et application du procede
US6656697B1 (en) * 1998-09-28 2003-12-02 Lifescan, Inc. Diagnostics based on tetrazolium compounds
TW496775B (en) * 1999-03-15 2002-08-01 Aviva Bioscience Corp Individually addressable micro-electromagnetic unit array chips
US6858439B1 (en) * 1999-03-15 2005-02-22 Aviva Biosciences Compositions and methods for separation of moieties on chips
US6511967B1 (en) * 1999-04-23 2003-01-28 The General Hospital Corporation Use of an internalizing transferrin receptor to image transgene expression
US6613581B1 (en) * 1999-08-26 2003-09-02 Caliper Technologies Corp. Microfluidic analytic detection assays, devices, and integrated systems
FR2798673B1 (fr) * 1999-09-16 2004-05-28 Exonhit Therapeutics Sa Methodes et compositions pour la detection d'evenements pathologiques
US6875619B2 (en) * 1999-11-12 2005-04-05 Motorola, Inc. Microfluidic devices comprising biochannels
US7346200B1 (en) * 1999-11-18 2008-03-18 Ikonisys, Inc. Method and apparatus for computer controlled cell based diagnosis
US6618679B2 (en) * 2000-01-28 2003-09-09 Althea Technologies, Inc. Methods for analysis of gene expression
MXPA03000506A (es) * 2000-07-18 2004-09-10 Correlogic Systems Inc Proceso para discriminiar entre estados biologicos basados en patrones escondidos de datos biologicos.
US20040005582A1 (en) * 2000-08-10 2004-01-08 Nanobiodynamics, Incorporated Biospecific desorption microflow systems and methods for studying biospecific interactions and their modulators
WO2002023163A1 (fr) * 2000-09-15 2002-03-21 California Institute Of Technology Dispositifs a debit transversal microfabriques et procedes associes
US20030007894A1 (en) * 2001-04-27 2003-01-09 Genoptix Methods and apparatus for use of optical forces for identification, characterization and/or sorting of particles
EP1368369A4 (fr) * 2000-11-15 2006-02-22 Hoffmann La Roche Methodes et reactifs permettant d'identifier des cellules embryonnaires rares dans le systeme circulatoire maternel
US20020155587A1 (en) * 2001-04-20 2002-10-24 Sequenom, Inc. System and method for testing a biological sample
FR2824144B1 (fr) * 2001-04-30 2004-09-17 Metagenex S A R L Methode de diagnostic prenatal sur cellule foetale isolee du sang maternel
US20020166760A1 (en) * 2001-05-11 2002-11-14 Prentiss Mara G. Micromagentic systems and methods for microfluidics
CA2466896A1 (fr) * 2001-09-06 2003-03-20 Adnagen Ag Procede et kit de diagnostic destines a la selection et/ou detection qualitative et/ou quantitative de cellules
DE10143776A1 (de) * 2001-09-06 2003-04-03 Adnagen Ag Verfahren und Kit zur Diagnostik oder Behandlungskontrolle von Brustkrebs
US7166443B2 (en) * 2001-10-11 2007-01-23 Aviva Biosciences Corporation Methods, compositions, and automated systems for separating rare cells from fluid samples
US20030178641A1 (en) * 2002-01-23 2003-09-25 Blair Steven M. Microfluidic platforms for use with specific binding assays, specific binding assays that employ microfluidics, and methods
WO2003069421A2 (fr) * 2002-02-14 2003-08-21 Immunivest Corporation Procedes et algorithmes pour le comptage de cellules dans un cytometre peu onereux
US7312085B2 (en) * 2002-04-01 2007-12-25 Fluidigm Corporation Microfluidic particle-analysis systems
US7141369B2 (en) * 2002-04-25 2006-11-28 Semibio Technology, Inc. Measuring cellular metabolism of immobilized cells
US7727720B2 (en) * 2002-05-08 2010-06-01 Ravgen, Inc. Methods for detection of genetic disorders
US7272252B2 (en) * 2002-06-12 2007-09-18 Clarient, Inc. Automated system for combining bright field and fluorescent microscopy
US20040018611A1 (en) * 2002-07-23 2004-01-29 Ward Michael Dennis Microfluidic devices for high gradient magnetic separation
US9435799B2 (en) * 2002-07-31 2016-09-06 Janssen Diagnostics, Inc. Methods and reagents for improved selection of biological materials
US7150834B2 (en) * 2003-07-31 2006-12-19 Arryx, Inc. Multiple laminar flow-based rate zonal or isopycnic separation with holographic optical trapping of blood cells and other static components
US20060008807A1 (en) * 2002-08-23 2006-01-12 O'hara Shawn M Multiparameter analysis of comprehensive nucleic acids and morphological features on the same sample
EP2359689B1 (fr) * 2002-09-27 2015-08-26 The General Hospital Corporation Dispositif microfluidique pour la séparation de cellules et usage du dispositif
WO2004037374A2 (fr) * 2002-10-23 2004-05-06 The Trustees Of Princeton University Procede de separation continue de particules faisant appel a des reseaux d'obstacles alignes de maniere asymetrique par rapport a des champs
AU2003291677A1 (en) * 2002-10-30 2004-05-25 Pointilliste, Inc. Methods for producing polypeptide-tagged collections and capture systems containing the tagged polypeptides
WO2004042033A2 (fr) * 2002-11-01 2004-05-21 The Board Of Trustees Of The Leland Stanford Junior University Cellules souches circulantes et utilisations en rapport
JPWO2004051231A1 (ja) * 2002-11-29 2006-04-06 日本電気株式会社 分離装置および分離方法
US20040197832A1 (en) * 2003-04-03 2004-10-07 Mor Research Applications Ltd. Non-invasive prenatal genetic diagnosis using transcervical cells
US20050221341A1 (en) * 2003-10-22 2005-10-06 Shimkets Richard A Sequence-based karyotyping
WO2005056838A1 (fr) * 2003-12-08 2005-06-23 The Clinic For Special Children Association de polymorphismes tspyl avec le syndrome siddt
US7468249B2 (en) * 2004-05-05 2008-12-23 Biocept, Inc. Detection of chromosomal disorders
US20060051265A1 (en) * 2004-09-08 2006-03-09 Health Research, Inc. Apparatus and method for sorting microstructures in a fluid medium
KR20060032466A (ko) * 2004-10-12 2006-04-17 삼성전자주식회사 이동 단말기의 핸드오버를 위한 광대역 무선 접속 통신시스템 및 그 시스템에서의 이동 단말기의 핸드오버 방법
US20060094109A1 (en) * 2004-11-02 2006-05-04 Immunivest Corporation Device and method for analytical cell imaging
EP2594631A1 (fr) * 2005-04-05 2013-05-22 Cellpoint Diagnostics Dispositifs et procédés détection de cellules tumorales circulantes et d'autres particules
US20070196820A1 (en) * 2005-04-05 2007-08-23 Ravi Kapur Devices and methods for enrichment and alteration of cells and other particles
US20070026418A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026417A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026414A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026413A1 (en) * 2005-07-29 2007-02-01 Mehmet Toner Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026415A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070037173A1 (en) * 2005-08-12 2007-02-15 Allard Jeffrey W Circulating tumor cells (CTC's): early assessment of time to progression, survival and response to therapy in metastatic cancer patients
US20070059680A1 (en) * 2005-09-15 2007-03-15 Ravi Kapur System for cell enrichment
US20070026419A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026416A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US8921102B2 (en) * 2005-07-29 2014-12-30 Gpb Scientific, Llc Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070059719A1 (en) * 2005-09-15 2007-03-15 Michael Grisham Business methods for prenatal Diagnosis
US20070059683A1 (en) * 2005-09-15 2007-03-15 Tom Barber Veterinary diagnostic system
US20070059781A1 (en) * 2005-09-15 2007-03-15 Ravi Kapur System for size based separation and analysis
US20070059718A1 (en) * 2005-09-15 2007-03-15 Mehmet Toner Systems and methods for enrichment of analytes
US20070059774A1 (en) * 2005-09-15 2007-03-15 Michael Grisham Kits for Prenatal Testing
US20070059716A1 (en) * 2005-09-15 2007-03-15 Ulysses Balis Methods for detecting fetal abnormality
EP1946114A4 (fr) * 2005-09-21 2010-05-26 Ccc Diagnostics Llc Procedures de test diagnostique exhaustives pour chimiotherapies anticancereuses personnalisees
JP5129149B2 (ja) * 2005-10-31 2013-01-23 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン 癌を処置および診断するための組成物および方法
EP1984030B1 (fr) * 2006-01-30 2013-05-08 The Scripps Research Institute Procedes de detection de cellules tumorales circulantes et procedes de diagnostic de cancer chez un sujet mammalien
EP3591068A1 (fr) * 2006-02-02 2020-01-08 The Board of Trustees of the Leland Stanford Junior University Dépistage génétique non invasif du f tus par analyse numérique
SI2351858T1 (sl) * 2006-02-28 2015-06-30 University Of Louisville Research Foundation Med Center Three, Zaznavanje fetalnih kromosomskih nenormalnosti z uporabo tandema polimorfizmov posameznih nukleotidov
WO2007112418A2 (fr) * 2006-03-28 2007-10-04 Baylor College Of Medicine Criblage du syndrome de down
US7735652B2 (en) * 2006-06-01 2010-06-15 The Trustees Of Princeton University Apparatus and method for continuous particle separation
US20080124721A1 (en) * 2006-06-14 2008-05-29 Martin Fuchs Analysis of rare cell-enriched samples
WO2008014516A2 (fr) * 2006-07-28 2008-01-31 Living Microsystems, Inc. Sélection de cellules au moyen de biomarqueurs
US20080113350A1 (en) * 2006-11-09 2008-05-15 Terstappen Leon W M M Blood test to monitor the genetic changes of progressive cancer using immunomagnetic enrichment and fluorescence in situ hybridization (FISH)
US8343440B2 (en) * 2009-03-27 2013-01-01 Seiko Epson Corporation Cell separating apparatus and cell separating method

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060073125A1 (en) * 2000-08-03 2006-04-06 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
WO2004113877A1 (fr) * 2003-06-13 2004-12-29 The General Hospital Corporation Systemes microfluidiques d'elimination basee sur la taille de globules rouges et de plaquettes du sang
US20060223178A1 (en) * 2005-04-05 2006-10-05 Tom Barber Devices and methods for magnetic enrichment of cells and other particles

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BODE ET AL.: "Mutations in the tyrosine kinase domain of the EGFR gene are rare in synovial sarcoma", MOD. PATHOL., vol. 19, no. 4, April 2005 (2005-04-01), pages 541 - 547 *
CHOESMEL ET AL.: "Enrichment methods to detect bone marrow micrometastases in breast carcinoma patients: clinical relevance", BREAST CANCER RES., vol. 6, no. 5, 29 July 2004 (2004-07-29), pages R556 - R570, XP021012062 *
KRIVACIC ET AL.: "A rare-cell detector for cancer", PROC. NATL. ACAD. SCI. USA, vol. 101, no. 29, 20 July 2004 (2004-07-20), pages 10501 - 10504, XP002432922 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
US11674176B2 (en) 2006-06-14 2023-06-13 Verinata Health, Inc Fetal aneuploidy detection by sequencing
US9493831B2 (en) 2010-01-23 2016-11-15 Verinata Health, Inc. Methods of fetal abnormality detection
US10718020B2 (en) 2010-01-23 2020-07-21 Verinata Health, Inc. Methods of fetal abnormality detection
US10324011B2 (en) 2013-03-15 2019-06-18 The Trustees Of Princeton University Methods and devices for high throughput purification
US10852220B2 (en) 2013-03-15 2020-12-01 The Trustees Of Princeton University Methods and devices for high throughput purification
US11142746B2 (en) 2013-03-15 2021-10-12 University Of Maryland, Baltimore High efficiency microfluidic purification of stem cells to improve transplants
US11486802B2 (en) 2013-03-15 2022-11-01 University Of Maryland, Baltimore Methods and devices for high throughput purification
US11493428B2 (en) 2013-03-15 2022-11-08 Gpb Scientific, Inc. On-chip microfluidic processing of particles
CN107208164A (zh) * 2015-02-13 2017-09-26 泊克玛科技有限公司 用于对循环肿瘤细胞进行测序的血液样品处理
US11268091B2 (en) 2018-12-13 2022-03-08 Dna Script Sas Direct oligonucleotide synthesis on cells and biomolecules

Also Published As

Publication number Publication date
US20160002737A1 (en) 2016-01-07
US20080124721A1 (en) 2008-05-29
WO2007147018A1 (fr) 2007-12-21
US20080090239A1 (en) 2008-04-17
US20130324418A1 (en) 2013-12-05

Similar Documents

Publication Publication Date Title
US11781187B2 (en) Rare cell analysis using sample splitting and DNA tags
US11261492B2 (en) Methods for the diagnosis of fetal abnormalities
US20080090239A1 (en) Rare cell analysis using sample splitting and dna tags
CA2655272C (fr) Analyse de cellules rares avec recours au fractionnement d'echantillons et a des marqueurs d'adn
EP3406736B1 (fr) Procédés pour le diagnostic d'anomalies fétales
AU2013204127B2 (en) Rare cell analysis using sample splitting and DNA tags

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07873985

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 07873985

Country of ref document: EP

Kind code of ref document: A1